1
|
Nguyen AM, Semeano A, Quach V, Inoue A, Nichols DE, Yano H. Characterization of Gα s and Gα olf activation by catechol and non-catechol dopamine D1 receptor agonists. iScience 2025; 28:112345. [PMID: 40384932 PMCID: PMC12084000 DOI: 10.1016/j.isci.2025.112345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 01/02/2025] [Accepted: 03/31/2025] [Indexed: 05/20/2025] Open
Abstract
The dopamine D1 receptor (D1R) couples to Gαs and Gαolf and is crucial in regulating neurological and neuropsychiatric functions. In the brain, Gαolf is predominantly found in the striatum whereas Gαs is expressed elsewhere. Our in vitro assays revealed that the tetracyclic catechol agonists dihydrexidine, methyl-dihydrexidine, doxanthrine, and the non-catechol compounds PF-8294, PF-6142 exerted full agonism for Gαs coupling but only partial agonism for Gαolf coupling. In contrast, the non-catechol agonist tavapadon acted as a full agonist at Gαolf and a partial agonist at Gαs. The effects of these ligands on the thalamocortical and striatonigral electrophysiological events, as well as on the locomotor activity and cognitive function of mice agreed with their selectivity profiles in vitro. These findings suggest the possibility of achieving region-specific pharmacology and open new directions for developing D1R drugs to treat relevant neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Anh Minh Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Ana Semeano
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Vianna Quach
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - David E. Nichols
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Hideaki Yano
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, MA, USA
| |
Collapse
|
2
|
Fonteneau C, Tamayo Z, Price A, Pan L, Afriyie-Agyemang Y, Agrawal S, Butler A, Cail C, Calkins M, Chakilam S, Forselius-Bielen K, Fram G, Frazier A, Gil R, Govil P, Gray DL, Grinband J, Gur RC, Haubold NK, Heffernan Z, Kegeles L, Kohler C, Lin C, Lu J, Mayer M, Pham P, Perlman G, Rahmati M, Ranganathan M, Santamauro NP, Schutte CT, Selloni A, Van Snellenberg J, Surti T, Wolf DH, Zharyy C, Abi-Dargham A, Gur RE, Lieberman JA, Kantrowitz JT, Anticevic A, Cho YT, Krystal JH. A Translational Neuroscience & Computational Evaluation of a D1R Partial Agonist for Schizophrenia (TRANSCENDS): Rationale and Study Design of a Brain-Based Clinical Trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.18.25326082. [PMID: 40321245 PMCID: PMC12047955 DOI: 10.1101/2025.04.18.25326082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Despite decades of research, cognitive impairment remains a critical untreated symptom for many patients with schizophrenia. One way to accelerate the development of pro-cognitive therapies for schizophrenia is to evaluate compounds using biomarker approaches tailored to relevant neural mechanisms. While D1/D5 receptor (D1R/D5R) agonism has been extensively studied in neuroscience, its therapeutic potential for cognitive impairment in schizophrenia remains untapped. The Translational Neuroscience & Computational Evaluation of a D1R Partial Agonist for Schizophrenia (TRANSCENDS) clinical trial tests this mechanism using a 'target engagement' approach. Multiple, double-blind doses of a D1/D5R partial agonist were administered in advance of a functional neuroimaging (fMRI) session that deployed a cognitive paradigm explicitly designed to capture a translational micro-circuit mechanism underlying spatial working memory in patients with schizophrenia. Specifically, this study will assess whether the D1R/D5R partial agonist CVL-562 induces a dose-dependent engagement of spatial working memory circuits in schizophrenia using fMRI. This design, and the use of spatial working memory neural circuits as a dependent measure, was selected on the basis of a translational and computational understanding of prefrontal micro-circuitry and a mechanistic understanding of the role of D1R/D5Rs in schizophrenia. To enhance data integration and scalability, TRANSCENDS employs an automated informatics framework for seamless neuroimaging data sharing and electronic clinical data capture. This ensures high-standards for regulatory compliance, data quality, and data sharing across sites, improving aspects of current clinical trial data management. We share the study design and approach with the goal of advancing future pro-cognitive drug development and strategies for developing mechanistically-driven biomarkers in psychiatry.
Collapse
Affiliation(s)
- Clara Fonteneau
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Zailyn Tamayo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Ally Price
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Lining Pan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Shriya Agrawal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Audrey Butler
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Courtney Cail
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Monica Calkins
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | - Geena Fram
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Allea Frazier
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Roberto Gil
- Department of Psychiatry, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | | | | | - Jack Grinband
- New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ruben C Gur
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Natalka K Haubold
- Department of Psychiatry, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Zachary Heffernan
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Larry Kegeles
- New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Christian Kohler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Chenyang Lin
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Jing Lu
- Investigational Drug Service, Yale New Haven Hospital, New Haven, CT, USA
| | - Megan Mayer
- New York State Psychiatric Institute, New York, NY
| | - Phuong Pham
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Greg Perlman
- Department of Psychiatry, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Masih Rahmati
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Mohini Ranganathan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Nicole P Santamauro
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Alexandria Selloni
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Jared Van Snellenberg
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Toral Surti
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Daniel H Wolf
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Catrin Zharyy
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Anissa Abi-Dargham
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Raquel E Gur
- Department of Psychiatry, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | | | - Joshua T Kantrowitz
- Department of Psychiatry, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
- Columbia University, College of Physicians and Surgeons, New York, NY
- Nathan Kline Institute, Orangeburg, NY, USA
| | - Alan Anticevic
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Youngsun T Cho
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - John H Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
3
|
Nilson AN, Felsing DE, Wang P, Jain MK, Zhou J, Allen JA. Functionally Selective Dopamine D1 Receptor Endocytosis and Signaling by Catechol and Noncatechol Agonists. Biochemistry 2025; 64:1572-1588. [PMID: 40111449 DOI: 10.1021/acs.biochem.4c00876] [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] [Indexed: 03/22/2025]
Abstract
The dopamine D1 receptor (D1R) has fundamental roles in voluntary movement and memory and is a validated drug target for neurodegenerative and neuropsychiatric disorders. However, previously developed D1R selective agonists possess a catechol moiety which displays poor pharmacokinetic properties. The first selective noncatechol D1R agonists were recently discovered and unexpectedly many of these ligands showed G protein biased signaling. Here, we investigate both catechol and noncatechol D1R agonists to validate potential biased signaling and examine if this impacts agonist-induced D1R endocytosis. We determined that most, but not all, noncatechol agonists display G protein biased signaling at the D1R and have reduced or absent β-arrestin2 recruitment. A notable exception was compound (Cmpd) 19, a noncatechol agonist with full efficacy at both D1R-G protein and D1R-β-arrestin2 pathways. In addition, the catechol ligand A-77636 was a highly potent, super agonist for D1R-β-arrestin2 activity. When examined for agonist-induced D1R endocytosis, balanced agonists SKF-81297 and Cmpd 19 induced robust D1R endocytosis while the G protein biased agonists did not. The β-arrestin2 super agonist, A-77636, showed statistically significant increases in D1R endocytosis. Moreover, β-arrestin2 recruitment efficacy of tested agonists strongly correlated with total D1R endocytosis. Taken together, these results indicate the degree of D1R signaling functional selectivity profoundly impacts D1R endocytosis regardless of pharmacophore. The range of functional selectivity of these D1R agonists will provide valuable tools to further investigate D1R signaling, trafficking and therapeutic potential.
Collapse
Affiliation(s)
- Ashley N Nilson
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
| | - Daniel E Felsing
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
| | - Pingyuan Wang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
- Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
| | - Manish K Jain
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
- Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
| | - John A Allen
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555-0615, United States
| |
Collapse
|
4
|
Fan L, Wang S. Biased GPCR Signaling: Possible Mechanisms and Therapeutic Applications. Biochemistry 2025; 64:1180-1192. [PMID: 40016120 DOI: 10.1021/acs.biochem.4c00827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Biased signaling refers to the phenomenon where a ligand selectively activates specific downstream pathways of G protein-coupled receptors (GPCRs), such as the G protein-mediated pathway or the β-arrestin-mediated pathway. This mechanism can be influenced by receptor bias, ligand bias, system bias and spatial bias, all of which are shaped by the receptor's conformational distinctions and kinetics. Since GPCRs are the largest class of drug targets, signaling bias garnered significant attention for its potential to enhance therapeutic efficacy while minimizing side effects. Despite intensive investigation, a major challenge lies in translating in vitro ligand efficacy into in vivo biological responses due to the dynamic and multifaceted nature of the in vivo environment. This review delves into the current understanding of GPCR-biased signaling, examining the role of structural bias at the molecular level, the impact of kinetic context on system and observational bias, and the challenges of applying these insights in drug development. It further explores future directions for advancing biased signaling applications, offering valuable perspectives on how to bridge the gap between in vitro studies and in vivo therapeutic design, ultimately accelerating the development of viable, biased therapeutics.
Collapse
Affiliation(s)
- Luyu Fan
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Sheng Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
5
|
De Risi M, Cavezza D, Torromino G, Capalbo A, Cundin XB, Di Martino R, Alvino FG, Iemolo A, Speranza L, Perrone-Capano C, Crispino M, Cirillo C, Luini A, Sacco F, Grumati P, De Leonibus E. Cortico-striatal circuit mechanisms drive the effects of D1 dopamine agonists on memory capacity in mice through cAMP/PKA signalling. Nat Commun 2025; 16:2615. [PMID: 40097401 PMCID: PMC11914583 DOI: 10.1038/s41467-025-57788-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
Working memory capacity (WMC), the number of items remembered in a short-time interval, is regulated by fronto-striatal dopamine (DA) and is reduced in schizophrenia. We investigated how excessive and insufficient D1 dopamine receptor stimulation impairs and expands WMC, focusing on the cAMP/PKA pathway in the fronto-striatal circuit. Low doses of the D1 agonist SKF 38393 enhance WMC by activating the striatum (mice remember more objects), while high doses, paradoxically, impair WMC, activating the same pathway in the medial prefrontal cortex (mPFC) but inhibiting it in the striatum. This impairment, arising from mPFC-driven recruitment of inhibitory striatal parvalbumin interneurons, can be prevented by optogenetic inhibition of the mPFC-striatal pathway. Low doses of SKF 38393 also rescue WMC deficits in a schizophrenia mouse model. These results highlight the need for a systems pharmacology approach that considers complex brain interactions and intracellular signalling pathways, rather than isolated drug-receptor interactions, to develop memory-enhancing treatments.
Collapse
MESH Headings
- Animals
- Receptors, Dopamine D1/agonists
- Receptors, Dopamine D1/metabolism
- Prefrontal Cortex/drug effects
- Prefrontal Cortex/metabolism
- Prefrontal Cortex/physiology
- Cyclic AMP/metabolism
- Schizophrenia/physiopathology
- Schizophrenia/metabolism
- Schizophrenia/drug therapy
- Memory, Short-Term/drug effects
- Memory, Short-Term/physiology
- Signal Transduction/drug effects
- Mice
- Dopamine Agonists/pharmacology
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Corpus Striatum/drug effects
- Corpus Striatum/metabolism
- Corpus Striatum/physiology
- Male
- 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology
- Mice, Inbred C57BL
- Optogenetics
- Interneurons/metabolism
- Interneurons/drug effects
- Disease Models, Animal
Collapse
Affiliation(s)
- Maria De Risi
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Monterotondo (Rome), Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli (Naples), Italy
| | - Diletta Cavezza
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Monterotondo (Rome), Italy
- PhD Program in Behavioral Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Giulia Torromino
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Monterotondo (Rome), Italy
- Department of Humanistic Studies, University of Naples Federico II, Naples, Italy
| | - Anita Capalbo
- Institute of Biochemistry and Cell Biology (IBBC), Naples, Italy
| | - Xabier Bujanda Cundin
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli (Naples), Italy
| | | | - Filomena Grazia Alvino
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Monterotondo (Rome), Italy
| | - Attilio Iemolo
- Institute of Genetics and Biophysics (IGB), National Research Council (CNR), Naples, Italy
| | - Luisa Speranza
- Institute of Genetics and Biophysics (IGB), National Research Council (CNR), Naples, Italy
| | - Carla Perrone-Capano
- Institute of Genetics and Biophysics (IGB), National Research Council (CNR), Naples, Italy
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Carmine Cirillo
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli (Naples), Italy
| | - Alberto Luini
- Institute of Biochemistry and Cell Biology (IBBC), Naples, Italy
| | - Francesca Sacco
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli (Naples), Italy
| | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli (Naples), Italy
| | - Elvira De Leonibus
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Monterotondo (Rome), Italy.
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli (Naples), Italy.
| |
Collapse
|
6
|
Kumar S, Rastogi SK, Roy S, Sharma K, Kumar S, Maity D, Chand D, Vishwakarma S, Gayen JR, Srivastava KR, Kumar R, Yadav PN. Discovery and structure - activity relationships of 2,4,5-trimethoxyphenyl pyrimidine derivatives as selective D5 receptor partial agonists. Bioorg Chem 2024; 153:107809. [PMID: 39270528 DOI: 10.1016/j.bioorg.2024.107809] [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: 07/15/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
Dopamine receptors are therapeutic targets for the treatment of various neurological and psychiatric disorders, including Parkinson's and Alzheimer's. Previously, PF-06649751 (tavapadon), PF-2562 and PW0464 have been discovered as potent and selective G protein-biased D1/D5 receptor agonists with optimal pharmacokinetic properties. However, no selective D5R agonist has been reported yet. In this context, we designed and synthesized forty non-catecholamines-based pyrimidine derivatives and identified four pyrimidine derivatives as selective D5R partial agonists. Using cAMP-based GloSensor assay in transiently transfected HEK293T cells with human D1 or D5 receptors, we discovered that compound 5c (4-(4-bromophenyl)-6-(2,4,5-trimethoxyphenyl)pyrimidin-2-amine) exhibited modest D5R agonist activity. This leads us to explore various modifications of this scaffold to improve the D5 agonist potency and efficacy. Using molecular docking, and rational design followed by their evaluation at D1 and D5 receptors for agonist activity, we identified three new derivatives, 5j, 5h, and 5e. The most potent compound of this series 5j (4-(4-iodophenyl)-6-(2,4,5-trimethoxyphenyl)pyrimidin-2-amine), exhibited EC50 of 269.7 ± 6.6 nM. Mice microsomal stability studies revealed that 5j is quite stable (>70 % at 1 hr). Furthermore, pharmacokinetic analysis of 5j (20 mg/kg, p.o) in C57BL/6j mice showed that 5j is readily absorbed via oral route of dosing and also enters into the brain (plasma Tmax: 1 h, Cmax: 51.10 ± 13.51 ng/ml; Brain Tmax: 0.5 h, Cmax: 22.54 ± 4.08 ng/ml). We further determined the in-vivo effect of 5j on cognition in scopolamine-induced amnesia in C57BL/6j mice. We observed that 5j (10 mg/kg, p.o) alleviated scopolamine-induced impairment in short-term memory and social recognition, which were blocked by D1/D5 antagonist SCH23390 (0.1 mg/kg, i.p.). Furthermore, 5j did not exhibit any cytotoxicity (up to 10 µM) or in vivo acute toxicity up to 200 mg/kg (p.o). These results strongly suggest that 5j could be further developed for treating neurological disorders wherein the D5 receptors play pivotal roles.
Collapse
Affiliation(s)
- Sakesh Kumar
- Neuroscience and Ageing Biology Division, CSIR-Central Drug Research Institute, Lucknow, U.P., (226031), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., (201002), India
| | - Sumit K Rastogi
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, U.P., (226031), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., (201002), India
| | - Subrata Roy
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, U.P., (226031), India
| | - Kajal Sharma
- Neuroscience and Ageing Biology Division, CSIR-Central Drug Research Institute, Lucknow, U.P., (226031), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., (201002), India
| | - Santosh Kumar
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, U.P., (226031), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., (201002), India
| | - Debalina Maity
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, U.P., (226031), India
| | - Diwan Chand
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, U.P., (226031), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., (201002), India
| | - Sachin Vishwakarma
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, U.P., (226031), India
| | - Jiaur R Gayen
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., (201002), India; Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, U.P., (226031), India
| | - Kinshuk R Srivastava
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, U.P., (226031), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., (201002), India
| | - Ravindra Kumar
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, U.P., (226031), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., (201002), India.
| | - Prem N Yadav
- Neuroscience and Ageing Biology Division, CSIR-Central Drug Research Institute, Lucknow, U.P., (226031), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., (201002), India.
| |
Collapse
|
7
|
Fan L, Zhuang Y, Wu H, Li H, Xu Y, Wang Y, He L, Wang S, Chen Z, Cheng J, Xu HE, Wang S. Structural basis of psychedelic LSD recognition at dopamine D 1 receptor. Neuron 2024; 112:3295-3310.e8. [PMID: 39094559 DOI: 10.1016/j.neuron.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/11/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024]
Abstract
Understanding the kinetics of LSD in receptors and subsequent induced signaling is crucial for comprehending both the psychoactive and therapeutic effects of LSD. Despite extensive research on LSD's interactions with serotonin 2A and 2B receptors, its behavior on other targets, including dopamine receptors, has remained elusive. Here, we present cryo-EM structures of LSD/PF6142-bound dopamine D1 receptor (DRD1)-legobody complexes, accompanied by a β-arrestin-mimicking nanobody, NBA3, shedding light on the determinants of G protein coupling versus β-arrestin coupling. Structural analysis unveils a distinctive binding mode of LSD in DRD1, particularly with the ergoline moiety oriented toward TM4. Kinetic investigations uncover an exceptionally rapid dissociation rate of LSD in DRD1, attributed to the flexibility of extracellular loop 2 (ECL2). Moreover, G protein can stabilize ECL2 conformation, leading to a significant slowdown in ligand's dissociation rate. These findings establish a solid foundation for further exploration of G protein-coupled receptor (GPCR) dynamics and their relevance to signal transduction.
Collapse
Affiliation(s)
- Luyu Fan
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Youwen Zhuang
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongyu Wu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Huiqiong Li
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Youwei Xu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yue Wang
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Licong He
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Shishan Wang
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang 261021, China
| | - Zhangcheng Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Lingang Laboratory, Shanghai 200031, China.
| | - Sheng Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
8
|
Roth BL, Krumm BE. Molecular glues as potential GPCR therapeutics. Biochem Pharmacol 2024; 228:116402. [PMID: 38945274 DOI: 10.1016/j.bcp.2024.116402] [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: 01/24/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
"Molecular Glues" are defined as small molecules that can either be endogenous or synthetic which promote interactions between proteins at their interface. Allosteric modulators, specifically GPCR allosteric modulators, can promote both the association and the dissociation of a given receptor's transducer but accomplishes this "at a distance" from the interface. However, recent structures of GPCR G protein complexes in the presence of allosteric modulators indicate that some GPCR allosteric modulators can act as "molecular glues" interacting with both the receptor and the transducer at the interface biasing transducer signaling in both a positive and negative manner depending on the transducer. Given these phenomena we discuss the implications for this class of allosteric modulators to be used as molecular tools and for future drug development.
Collapse
Affiliation(s)
- Bryan L Roth
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Brian E Krumm
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
9
|
Rodríguez Rondón AV, Welling MS, van den Akker ELT, van Rossum EFC, Boon EMJ, van Haelst MM, Delhanty PJD, Visser JA. MC4R Variants Modulate α-MSH and Setmelanotide Induced Cellular Signaling at Multiple Levels. J Clin Endocrinol Metab 2024; 109:2452-2466. [PMID: 38567654 PMCID: PMC11403317 DOI: 10.1210/clinem/dgae210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/10/2024] [Indexed: 04/04/2024]
Abstract
CONTEXT The melanocortin-4 receptor (MC4R) plays an important role in body weight regulation. Pathogenic MC4R variants are the most common cause of monogenic obesity. OBJECTIVE We have identified 17 MC4R variants in adult and pediatric patients with obesity. Here we aimed to functionally characterize these variants by analyzing 4 different aspects of MC4R signaling. In addition, we aimed to analyze the effect of setmelanotide, a potent MC4R agonist, on these MC4R variants. MATERIALS AND METHODS Cell surface expression and α-melanocyte stimulating hormone (α-MSH)- or setmelanotide-induced cAMP response, β-arrestin-2 recruitment, and ERK activation were measured in cells expressing either wild type or variant MC4R. RESULTS We found a large heterogeneity in the function of these variants. We identified variants with a loss of response for all studied MC4R signaling, variants with no cAMP accumulation or ERK activation but normal β-arrestin-2 recruitment, and variants with normal cAMP accumulation and ERK activation but decreased β-arrestin-2 recruitment, indicating disrupted desensitization and signaling mechanisms. Setmelanotide displayed a greater potency and similar efficacy as α-MSH and induced significantly increased maximal cAMP responses of several variants compared to α-MSH. Despite the heterogeneity in functional response, there was no apparent difference in the obesity phenotype in our patients. CONCLUSION We show that these obesity-associated MC4R variants affect MC4R signaling differently yet lead to a comparable clinical phenotype. Our results demonstrate the clinical importance of assessing the effect of MC4R variants on a range of molecular signaling mechanisms to determine their association with obesity, which may aid in improving personalized treatment.
Collapse
Affiliation(s)
- Alejandra V Rodríguez Rondón
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Mila S Welling
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Pediatrics, Division of Endocrinology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Erica L T van den Akker
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Pediatrics, Division of Endocrinology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Elisabeth F C van Rossum
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Elles M J Boon
- Department of Human Genetics, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Mieke M van Haelst
- Department of Human Genetics, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Patric J D Delhanty
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Jenny A Visser
- Obesity Center CGG and Expertise Center Genetic Obesity, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
10
|
Dorogan M, Namballa HK, Harding WW. Natural Product-Inspired Dopamine Receptor Ligands. J Med Chem 2024; 67:12463-12484. [PMID: 39038276 PMCID: PMC11320586 DOI: 10.1021/acs.jmedchem.4c00537] [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: 03/03/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024]
Abstract
Due to their evolutionary bias as ligands for biologically relevant drug targets, natural products offer a unique opportunity as lead compounds in drug discovery. Given the involvement of dopamine receptors in various physiological and behavioral functions, they are linked to numerous diseases and disorders such as Parkinson's disease, schizophrenia, and substance use disorders. Consequently, ligands targeting dopamine receptors hold considerable therapeutic and investigative promise. As this perspective will highlight, dopamine receptor targeting natural products play a pivotal role as scaffolds with unique and beneficial pharmacological properties, allowing for natural product-inspired drug design and lead optimization. As such, dopamine receptor targeting natural products still have untapped potential to aid in the treatment of disorders and diseases related to central nervous system (CNS) and peripheral nervous system (PNS) dysfunction.
Collapse
Affiliation(s)
- Michael Dorogan
- Department
of Chemistry, Hunter College, City University
of New York, 695 Park
Avenue, New York, New York 10065, United States
| | - Hari K. Namballa
- Department
of Chemistry, Hunter College, City University
of New York, 695 Park
Avenue, New York, New York 10065, United States
| | - Wayne W. Harding
- Department
of Chemistry, Hunter College, City University
of New York, 695 Park
Avenue, New York, New York 10065, United States
- Program
in Biochemistry, CUNY Graduate Center, 365 Fifth Avenue, New York, New York 10016, United States
- Program
in Chemistry, CUNY Graduate Center, 365 Fifth Avenue, New York, New York 10016, United
States
| |
Collapse
|
11
|
Murphy RE, Wang P, Ali S, Smith HR, Felsing DE, Chen H, Zhou J, Allen JA. Discovery of 3-((4-Benzylpyridin-2-yl)amino)benzamides as Potent GPR52 G Protein-Biased Agonists. J Med Chem 2024; 67:9709-9730. [PMID: 38788241 PMCID: PMC11441106 DOI: 10.1021/acs.jmedchem.4c00856] [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] [Indexed: 05/26/2024]
Abstract
Orphan GPR52 is emerging as a promising neurotherapeutic target. Optimization of previously reported lead 4a employing an iterative drug design strategy led to the identification of a series of unique GPR52 agonists, such as 10a (PW0677), 15b (PW0729), and 24f (PW0866), with improved potency and efficacy. Intriguingly, compounds 10a and 24f showed greater bias for G protein/cAMP signaling and induced significantly less in vitro desensitization than parent compound 4a, indicating that reducing GPR52 β-arrestin activity with biased agonism results in sustained GPR52 activation. Further exploration of compounds 15b and 24f indicated improved potency and efficacy, and excellent target selectivity, but limited brain exposure warranting further optimization. These balanced and biased GPR52 agonists provide important pharmacological tools to study GPR52 activation, signaling bias, and therapeutic potential for neuropsychiatric and neurological diseases.
Collapse
Affiliation(s)
- Ryan E. Murphy
- Center for Addiction Sciences and Therapeutics, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Pingyuan Wang
- Center for Addiction Sciences and Therapeutics, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States; Present Address: Ocean University of China, Qingdao, Shangdong 266003, China
| | - Saghir Ali
- Center for Addiction Sciences and Therapeutics, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Hudson R. Smith
- Center for Addiction Sciences and Therapeutics, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Daniel E. Felsing
- Center for Addiction Sciences and Therapeutics, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States; Present Address: Neurocrine Biosciences, San Diego, California 92130, United States
| | - Haiying Chen
- Center for Addiction Sciences and Therapeutics, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Center for Addiction Sciences and Therapeutics, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - John A. Allen
- Center for Addiction Sciences and Therapeutics, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
12
|
Girmaw F. Review on allosteric modulators of dopamine receptors so far. Health Sci Rep 2024; 7:e1984. [PMID: 38505681 PMCID: PMC10948587 DOI: 10.1002/hsr2.1984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/21/2024] Open
Abstract
Background Contemporary research is predominantly directed towards allosteric modulators, a class of compounds designed to interact with specific sites distinct from the orthosteric site on G protein-coupled receptors. These allosteric modulators play a pivotal role in influencing diverse pharmacological effects, such as agonism/inverse agonism, efficacy modulation, and affinity modulation. One particularly intriguing aspect is the demonstrated capacity of allosteric modulation to enhance drug selectivity for therapeutic purposes, potentially leading to a reduction in serious side effects associated with traditional approaches. Allosteric ligands, a majority of which fall into the categories of negative allosteric modulators or positive allosteric modulators, exhibit the unique ability to either diminish or enhance the effects of endogenous ligands. Negative allosteric modulators weaken the response, while positive allosteric modulators intensify it. Additionally, silent allosteric modulators represent a distinct class that neither activates nor blocks the effects of endogenous ligands, adding complexity to the spectrum of allosteric modulation. In the broader context of central nervous system disorders, allosteric modulation takes center stage, particularly in the realm of dopamine receptors specifically, D1, D2, and D3 receptors. These receptors hold immense therapeutic potential for a range of conditions spanning neurodegenerative disorders to neurobehavioral and psychiatric disorders. The intricate modulation of dopamine receptors through allosteric mechanisms offers a nuanced and versatile approach to drug development. As research endeavors continue to unfold, the exploration of allosteric modulation stands as a promising frontier, holding the potential to reshape the landscape of drug discovery and therapeutic interventions in the field of neurology and psychiatry.
Collapse
Affiliation(s)
- Fentaw Girmaw
- Department of Pharmacy, College of Health ScienceWoldia UniversityWoldiaEthiopia
| |
Collapse
|
13
|
Bezard E, Gray D, Kozak R, Leoni M, Combs C, Duvvuri S. Rationale and Development of Tavapadon, a D1/D5-Selective Partial Dopamine Agonist for the Treatment of Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:476-487. [PMID: 36999711 PMCID: PMC10909821 DOI: 10.2174/1871527322666230331121028] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 04/01/2023]
Abstract
Currently, available therapeutics for the treatment of Parkinson's disease (PD) fail to provide sustained and predictable relief from motor symptoms without significant risk of adverse events (AEs). While dopaminergic agents, particularly levodopa, may initially provide strong motor control, this efficacy can vary with disease progression. Patients may suffer from motor fluctuations, including sudden and unpredictable drop-offs in efficacy. Dopamine agonists (DAs) are often prescribed during early-stage PD with the expectation they will delay the development of levodopa-associated complications, but currently available DAs are less effective than levodopa for the treatment of motor symptoms. Furthermore, both levodopa and DAs are associated with a significant risk of AEs, many of which can be linked to strong, repeated stimulation of D2/D3 dopamine receptors. Targeting D1/D5 dopamine receptors has been hypothesized to produce strong motor benefits with a reduced risk of D2/D3-related AEs, but the development of D1-selective agonists has been previously hindered by intolerable cardiovascular AEs and poor pharmacokinetic properties. There is therefore an unmet need in PD treatment for therapeutics that provide sustained and predictable efficacy, with strong relief from motor symptoms and reduced risk of AEs. Partial agonism at D1/D5 has shown promise for providing relief from motor symptoms, potentially without the AEs associated with D2/D3-selective DAs and full D1/D5-selective DAs. Tavapadon is a novel oral partial agonist that is highly selective at D1/D5 receptors and could meet these criteria. This review summarizes currently available evidence of tavapadon's therapeutic potential for the treatment of early through advanced PD.
Collapse
Affiliation(s)
- Erwan Bezard
- Université de Bordeaux, CNRS Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- Motac Neuroscience, Manchester, United Kingdom
| | | | | | | | | | | |
Collapse
|
14
|
Namballa HK, Decker AM, Dorogan M, Gudipally A, Goclon J, Harding WW. Fluoroalkoxylated C-3 and C-9 (S)-12-bromostepholidine analogues with D1R antagonist activity. Bioorg Chem 2023; 141:106862. [PMID: 37722267 PMCID: PMC10872833 DOI: 10.1016/j.bioorg.2023.106862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 09/20/2023]
Abstract
To illuminate the tolerance of fluoroalkoxylated groups at the C-3 and C-9 positions of tetrahydroprotoberberines (THPBs) on D1R activity, C-3 and C-9 fluoroalkoxylated analogues of (S)-12-bromostepholidine were prepared and evaluated. All compounds examined were D1R antagonists as measured by a cAMP assay. Our structure-activity studies herein indicate that the C-3 position tolerates a 1,1-difluoroethoxy substituent for D1R antagonist activity. Compound 13a was the most potent cAMP-based D1R antagonist identified and was also found to antagonize β-arrestin translocation in a TANGO assay. Affinity assessments at other dopamine receptors revealed that 13a is selective for D1R and unlike other naturally-occurring THPBs such as (S)-stepholidine, lacks D2R affinity. In preliminary biopharmaceutical assays, excellent BBB permeation was observed for 13a. Further pharmacological studies are warranted on (S)-stepholidine congeners to harvest their potential as a source of novel, druggable D1R-targeted agents.
Collapse
Affiliation(s)
- Hari K Namballa
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States
| | - Ann M Decker
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - Michael Dorogan
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States
| | - Ashok Gudipally
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States; Program in Chemistry, CUNY Graduate Center 365 5th Avenue, New York, NY 10016, United States
| | - Jakub Goclon
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States
| | - Wayne W Harding
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, United States; Program in Chemistry, CUNY Graduate Center 365 5th Avenue, New York, NY 10016, United States; Program in Biochemistry, CUNY Graduate Center 365 5th Avenue, New York, NY 10016, United States.
| |
Collapse
|
15
|
Isaacson SH, Hauser RA, Pahwa R, Gray D, Duvvuri S. Dopamine agonists in Parkinson's disease: Impact of D1-like or D2-like dopamine receptor subtype selectivity and avenues for future treatment. Clin Park Relat Disord 2023; 9:100212. [PMID: 37497384 PMCID: PMC10366643 DOI: 10.1016/j.prdoa.2023.100212] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 07/28/2023] Open
Abstract
Dopamine agonists (DAs) have demonstrated efficacy for the treatment of Parkinson's disease (PD) but are limited by adverse effects (AEs). DAs can vary considerably in their receptor subtype selectivity and affinity, chemical composition, receptor occupancy, and intrinsic activity on the receptor. Most currently approved DAs for PD treatment primarily target D2/D3 (D2-like) dopamine receptors. However, selective activation of D1/D5 (D1-like) dopamine receptors may enable robust activation of motor function while avoiding AEs related to D2/D3 receptor agonism. Full D1/D5 receptor-selective agonists have been explored in small, early-phase clinical studies, and although their efficacy for motor symptoms was robust, challenges with pharmacokinetics, bioavailability, cardiovascular AEs, and dyskinesia rates similar to levodopa prevented clinical advancement. Generally, repeated dopaminergic stimulation with full DAs is associated with frontostriatal dysfunction and sensitization that may induce plastic changes in the motor system, and neuroadaptations that produce long-term motor and nonmotor complications, respectively. Recent preclinical and clinical studies suggest that a D1/D5 receptor-selective partial agonist may hold promise for providing sustained, predictable, and robust motor control, while reducing risk for motor complications (e.g., levodopa-induced dyskinesia) and nonmotor AEs (e.g., impulse control disorders and excessive daytime sleepiness). Clinical trials are ongoing to evaluate this hypothesis. The potential emerging availability of novel dopamine receptor agonists with selective dopamine receptor pharmacology suggests that the older terminology "dopamine agonist" may need revision to distinguish older-generation D2/D3-selective agonists from D1/D5-selective agonists with distinct efficacy and tolerability characteristics.
Collapse
Affiliation(s)
- Stuart H. Isaacson
- Parkinson's Disease and Movement Disorders Center of Boca Raton, Boca Raton, FL, USA
| | - Robert A. Hauser
- Parkinson's Disease and Movement Disorders Center, Parkinson Foundation Center of Excellence, University of South Florida, Tampa, FL, USA
| | - Rajesh Pahwa
- Parkinson's Disease and Movement Disorder Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - David Gray
- Vigil Neuroscience, Inc, Watertown, MA, USA
| | | |
Collapse
|
16
|
Lewis MM, Van Scoy LJ, De Jesus S, Hakun JG, Eslinger PJ, Fernandez-Mendoza J, Kong L, Yang Y, Snyder BL, Loktionova N, Duvvuri S, Gray DL, Huang X, Mailman RB. Dopamine D 1 Agonists: First Potential Treatment for Late-Stage Parkinson's Disease. Biomolecules 2023; 13:829. [PMID: 37238699 PMCID: PMC10216182 DOI: 10.3390/biom13050829] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/17/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Current pharmacotherapy has limited efficacy and/or intolerable side effects in late-stage Parkinson's disease (LsPD) patients whose daily life depends primarily on caregivers and palliative care. Clinical metrics inadequately gauge efficacy in LsPD patients. We explored if a D1/5 dopamine agonist would have efficacy in LsPD using a double-blind placebo-controlled crossover phase Ia/b study comparing the D1/5 agonist PF-06412562 to levodopa/carbidopa in six LsPD patients. Caregiver assessment was the primary efficacy measure because caregivers were with patients throughout the study, and standard clinical metrics inadequately gauge efficacy in LsPD. Assessments included standard quantitative scales of motor function (MDS-UPDRS-III), alertness (Glasgow Coma and Stanford Sleepiness Scales), and cognition (Severe Impairment and Frontal Assessment Batteries) at baseline (Day 1) and thrice daily during drug testing (Days 2-3). Clinicians and caregivers completed the clinical impression of change questionnaires, and caregivers participated in a qualitative exit interview. Blinded triangulation of quantitative and qualitative data was used to integrate findings. Neither traditional scales nor clinician impression of change detected consistent differences between treatments in the five participants who completed the study. Conversely, the overall caregiver data strongly favored PF-06412562 over levodopa in four of five patients. The most meaningful improvements converged on motor, alertness, and functional engagement. These data suggest for the first time that there can be useful pharmacological intervention in LsPD patients using D1/5 agonists and also that caregiver perspectives with mixed method analyses may overcome limitations using methods common in early-stage patients. The results encourage future clinical studies and understanding of the most efficacious signaling properties of a D1 agonist for this population.
Collapse
Affiliation(s)
- Mechelle M. Lewis
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Translational Brain Research Center, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Lauren J. Van Scoy
- Translational Brain Research Center, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Hershey, PA 17033, USA
- Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Humanities, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Sol De Jesus
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Jonathan G. Hakun
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Paul J. Eslinger
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Radiology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Julio Fernandez-Mendoza
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Psychiatry, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Lan Kong
- Translational Brain Research Center, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Hershey, PA 17033, USA
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Yang Yang
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Translational Brain Research Center, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Bethany L. Snyder
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Translational Brain Research Center, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Natalia Loktionova
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | - Xuemei Huang
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Translational Brain Research Center, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Hershey, PA 17033, USA
- Department of Radiology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Kinesiology, Pennsylvania State University, University Park, PA 16802, USA
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Richard B. Mailman
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Translational Brain Research Center, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
17
|
Li H, Urs NM, Horenstein N. Computational insights into ligand-induced G protein and β-arrestin signaling of the dopamine D1 receptor. J Comput Aided Mol Des 2023; 37:227-244. [PMID: 37060492 DOI: 10.1007/s10822-023-00503-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
The dopamine D1 receptor (D1R), is a class A G protein coupled-receptor (GPCR) which has been a promising drug target for psychiatric and neurological disorders such as Parkinson's disease (PD). Previous studies have suggested that therapeutic effects can be realized by targeting the β-arrestin signaling pathway of dopamine receptors, while overactivation of the G protein-dependent pathways leads to side effects, such as dyskinesias. Therefore, it is highly desirable to develop a D1R ligand that selectively regulates the β-arrestin pathway. Currently, most D1R agonists are signaling-balanced and stimulate both G protein and β-arrestin pathways, with a few reports of G protein biased ligands. However, identification and characterization of β-arrestin biased D1R agonists has been a challenge thus far. In this study, we implemented Gaussian accelerated molecular dynamics (GaMD) simulations to provide valuable computational insights into the possible underlying molecular mechanism of the different signaling properties of two catechol and two non-catechol D1R agonists that are either G protein biased or signaling-balanced. Dynamic network analysis further identified critical residues in the allosteric signaling network of D1R for each ligand at different conformational or binding states. Some of these residues are crucial for G protein or arrestin signals of GPCRs based on previous studies. Finally, we provided a molecular design strategy which can be utilized by medicinal chemists to develop potential β-arrestin biased D1R ligands. The proposed hypotheses are experimentally testable and can guide the development of safer and more effective medications for a variety of CNS disorders.
Collapse
Affiliation(s)
- Haoxi Li
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Nikhil M Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, 32610, USA
| | - Nicole Horenstein
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
18
|
Neumann J, Hofmann B, Dhein S, Gergs U. Role of Dopamine in the Heart in Health and Disease. Int J Mol Sci 2023; 24:ijms24055042. [PMID: 36902474 PMCID: PMC10003060 DOI: 10.3390/ijms24055042] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Dopamine has effects on the mammalian heart. These effects can include an increase in the force of contraction, and an elevation of the beating rate and the constriction of coronary arteries. Depending on the species studied, positive inotropic effects were strong, very modest, or absent, or even negative inotropic effects occurred. We can discern five dopamine receptors. In addition, the signal transduction by dopamine receptors and the regulation of the expression of cardiac dopamine receptors will be of interest to us, because this might be a tempting area of drug development. Dopamine acts in a species-dependent fashion on these cardiac dopamine receptors, but also on cardiac adrenergic receptors. We will discuss the utility of drugs that are currently available as tools to understand cardiac dopamine receptors. The molecule dopamine itself is present in the mammalian heart. Therefore, cardiac dopamine might act as an autocrine or paracrine compound in the mammalian heart. Dopamine itself might cause cardiac diseases. Moreover, the cardiac function of dopamine and the expression of dopamine receptors in the heart can be altered in diseases such as sepsis. Various drugs for cardiac and non-cardiac diseases are currently in the clinic that are, at least in part, agonists or antagonists at dopamine receptors. We define the research needs in order to understand dopamine receptors in the heart better. All in all, an update on the role of dopamine receptors in the human heart appears to be clinically relevant, and is thus presented here.
Collapse
Affiliation(s)
- Joachim Neumann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, 06097 Halle, Germany
- Correspondence: ; Tel.: +49-345-557-1686; Fax: +49-345-557-1835
| | - Britt Hofmann
- Herzchirurgie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, 06097 Halle, Germany
| | - Stefan Dhein
- Medizinische Fakultät, Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, 04107 Leipzig, Germany
| | - Ulrich Gergs
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, 06097 Halle, Germany
| |
Collapse
|
19
|
Chen Q, Zhou X, Rehmel J, Steele JP, Svensson KA, Beck JP, Hembre EJ, Hao J. Ensemble Docking Approach to Mitigate Pregnane X Receptor-Mediated CYP3A4 Induction Risk. J Chem Inf Model 2023; 63:173-186. [PMID: 36473234 DOI: 10.1021/acs.jcim.2c01175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three structurally closely related dopamine D1 receptor positive allosteric modulators (D1 PAMs) based on a tetrahydroisoquinoline (THIQ) scaffold were profiled for their CYP3A4 induction potentials. It was found that the length of the linker at the C5 position greatly affected the potentials of these D1 PAMs as CYP3A4 inducers, and the level of induction correlated well with the activation of the pregnane X receptor (PXR). Based on the published PXR X-ray crystal structures, we built a binding model specifically for these THIQ-scaffold-based D1 PAMs in the PXR ligand-binding pocket via an ensemble docking approach and found the model could explain the observed CYP induction disparity. Combined with our previously reported D1 receptor homology model, which identified the C5 position as pointing toward the solvent-exposed space, our PXR-binding model coincidentally suggested that structural modifications at the C5 position could productively modulate the CYP induction potential while maintaining the D1 PAM potency of these THIQ-based PAMs.
Collapse
Affiliation(s)
- Qi Chen
- Discovery Chemistry Research and Technologies, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Xin Zhou
- Drug Disposition, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, California92121, United States
| | - Jessica Rehmel
- Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - James P Steele
- Quantitative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Kjell A Svensson
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - James P Beck
- Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, California92121, United States
| | - Erik J Hembre
- Discovery Chemistry Research and Technologies, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Junliang Hao
- Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, California92121, United States
| |
Collapse
|
20
|
Jing XZ, Yang HJ, Taximaimaiti R, Wang XP. Advances in the Therapeutic Use of Non-Ergot Dopamine Agonists in the Treatment of Motor and Non-Motor Symptoms of Parkinson's Disease. Curr Neuropharmacol 2023; 21:1224-1240. [PMID: 36111769 PMCID: PMC10286583 DOI: 10.2174/1570159x20666220915091022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/25/2022] [Accepted: 07/18/2022] [Indexed: 11/22/2022] Open
Abstract
Dopamine (DA) agonists, as an excellent dopamine replacement therapy for patients with early and advanced Parkinson's disease (PD), play a vital role in controlling motor and several nonmotor symptoms. Besides, the application of DA agonists may delay levodopa therapy and the associated risk of motor complications. Indeed, each DA agonist has unique pharmacokinetic and pharmacodynamic characteristics and therefore has different therapeutic efficacy and safety profile. The comorbidities, significant non-motor manifestations, concomitant medications, and clinical features of PD individuals should guide the selection of a specific DA agonist to provide a more patient-tailored treatment option. Thorough knowledge of DA agonists helps clinicians better balance clinical efficacy and side effects. Therefore, this review refers to recent English-written articles on DA agonist therapy for PD patients and summarizes the latest findings on non-ergot DA agonists as well as the advantages and disadvantages of each compound to help clinicians in the selection of a specific DA agonist. In addition, novel D1/D5 partial agonists and new formulations of DA agonists are also discussed.
Collapse
Affiliation(s)
- Xiao-Zhong Jing
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Department of Neurology, TongRen Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Hui-Jia Yang
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
| | - Reyisha Taximaimaiti
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xiao-Ping Wang
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Department of Neurology, TongRen Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Further studies on C2′-substituted 1-phenylbenzazepines as dopamine D1 receptor ligands. Bioorg Chem 2022; 127:105953. [DOI: 10.1016/j.bioorg.2022.105953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 11/20/2022]
|
22
|
Yang Y, Lewis MM, Kong L, Mailman RB. A Dopamine D 1 Agonist Versus Methylphenidate in Modulating Prefrontal Cortical Working Memory. J Pharmacol Exp Ther 2022; 382:88-99. [PMID: 35661631 PMCID: PMC9341252 DOI: 10.1124/jpet.122.001215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022] Open
Abstract
Methylphenidate is used widely to treat symptoms of attention-deficit/hyperactivity disorder (ADHD), but like other stimulants has significant side effects. This study used a rodent model (spontaneously hypertensive rat) of spatial working memory (sWM) to compare the effects of methylphenidate with the novel dopamine D1-like receptor agonist 2-methyldihydrexidine. Acute oral administration of methylphenidate (1.5 mg/kg) caused sWM improvement in half of the tested rats, but impairment in the others. Both improvement or impairment were eliminated by administration of the D1 antagonist SCH39266 directly into the prefrontal cortex (PFC). Conversely, 2-methyldihydrexidine showed greater sWM improvement compared with methylphenidate without significant impairment in any subject. Its effects correlated negatively with vehicle-treated baseline performance (i.e., rats with lower baseline performance improved more than rats with higher baseline performance). These behavioral effects were associated with neural activities in the PFC. Single neuron firing rate was changed, leading to the alteration in neuronal preference to correct or error behavioral responses. Overall, 2-methyldihydrexidine was superior to methylphenidate in decreasing the neuronal preference, prospectively, in the animals whose behavior was improved. In contrast, methylphenidate, but not 2-methyldihydrexidine, significantly decreased neuronal preference, retrospectively, in those animals who had impaired performance. These results suggest that a D1 agonist may be more effective than methylphenidate in regulating sWM-related behavior through neural modulation of the PFC, and thus may be superior to methylphenidate or other stimulants as ADHD pharmacotherapy. SIGNIFICANCE STATEMENT: Methylphenidate is effective in ADHD by its indirect agonist stimulation of dopamine and/or adrenergic receptors, but the precise effects on specific targets are unclear. This study compared methylphenidate to a dopamine D1 receptor-selective agonist by investigating effects on working memory occurring via neural modulation in the prefrontal cortex. The data suggest that pharmacological treatment selectively targeting the dopamine D1 may offer a superior approach to ADHD pharmacotherapy.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacology (Y.Y., M.M.L., R.B.M.), Department of Neurology (M.M.L., R.B.M.), and Department of Public Health Sciences (L.K.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Mechelle M Lewis
- Department of Pharmacology (Y.Y., M.M.L., R.B.M.), Department of Neurology (M.M.L., R.B.M.), and Department of Public Health Sciences (L.K.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Lan Kong
- Department of Pharmacology (Y.Y., M.M.L., R.B.M.), Department of Neurology (M.M.L., R.B.M.), and Department of Public Health Sciences (L.K.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Richard B Mailman
- Department of Pharmacology (Y.Y., M.M.L., R.B.M.), Department of Neurology (M.M.L., R.B.M.), and Department of Public Health Sciences (L.K.), Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
23
|
Zhong Z, He X, Ge J, Zhu J, Yao C, Cai H, Ye XY, Xie T, Bai R. Discovery of small-molecule compounds and natural products against Parkinson's disease: Pathological mechanism and structural modification. Eur J Med Chem 2022; 237:114378. [DOI: 10.1016/j.ejmech.2022.114378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/08/2021] [Accepted: 04/09/2022] [Indexed: 11/24/2022]
|
24
|
Yang Y, Lewis MM, Huang X, Dokholyan NV, Mailman RB. Dopamine D 1 receptor-mediated β-arrestin signaling: Insight from pharmacology, biology, behavior, and neurophysiology. Int J Biochem Cell Biol 2022; 148:106235. [PMID: 35688404 PMCID: PMC10266066 DOI: 10.1016/j.biocel.2022.106235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 02/16/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022]
Abstract
The awareness of the potential importance of functional selectivity/biased signaling has led to the discovery of biased compounds as both research tools and novel drugs. A major pan-receptor focus has been to identify GPCR-selective ligands that have bias in G protein-dependent vs. β-arrestin related signaling. Although this field has exploded during the past two decades, it is only recently that highly β-arrestin biased ligands for the dopamine D1 receptor were reported. We now summarize important pharmacological, molecular, and cellular studies relevant to D1-mediated β-arrestin-related signaling. It is intriguing that many results emerged from behavioral and physiological studies implying that bias toward or against D1-mediated β-arrestin either can improve or impair functional outcomes. We discuss the importance of understanding the translatability of cell and animal models to have more precise functional targeting to harness the value of this signaling pathway.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Translational Brain Research Center, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA.
| | - Mechelle M Lewis
- Department of Pharmacology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Department of Neurology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Translational Brain Research Center, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA
| | - Xuemei Huang
- Department of Pharmacology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Department of Neurology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Department of Humanities, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Department of Radiology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Department of Neurosurgery, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Department of Kinesiology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Translational Brain Research Center, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA
| | - Richard B Mailman
- Department of Pharmacology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Department of Neurology, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA; Translational Brain Research Center, Penn State Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
25
|
Teng X, Chen S, Nie Y, Xiao P, Yu X, Shao Z, Zheng S. Ligand recognition and biased agonism of the D1 dopamine receptor. Nat Commun 2022; 13:3186. [PMID: 35676276 PMCID: PMC9177848 DOI: 10.1038/s41467-022-30929-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 05/25/2022] [Indexed: 12/22/2022] Open
Abstract
Dopamine receptors are widely distributed in the central nervous system and are important therapeutic targets for treatment of various psychiatric and neurological diseases. Here, we report three cryo-electron microscopy structures of the D1 dopamine receptor (D1R)-Gs complex bound to two agonists, fenoldopam and tavapadon, and a positive allosteric modulator LY3154207. The structure reveals unusual binding of two fenoldopam molecules, one to the orthosteric binding pocket (OBP) and the other to the extended binding pocket (EBP). In contrast, one elongated tavapadon molecule binds to D1R, extending from OBP to EBP. Moreover, LY3154207 stabilizes the second intracellular loop of D1R in an alpha helical conformation to efficiently engage the G protein. Through a combination of biochemical, biophysical and cellular assays, we further show that the broad conformation stabilized by two fenoldopam molecules and interaction between TM5 and the agonist are important for biased signaling of D1R.
Collapse
Affiliation(s)
- Xiao Teng
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
- National Institute of Biological Sciences, Beijing, China
| | - Sijia Chen
- National Institute of Biological Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Yingying Nie
- National Institute of Biological Sciences, Beijing, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sanduo Zheng
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
- National Institute of Biological Sciences, Beijing, China.
- Graduate School of Peking Union Medical College, Beijing, China.
| |
Collapse
|
26
|
Li H, Mirabel R, Zimmerman J, Ghiviriga I, Phidd DK, Horenstein N, Urs NM. Structure-Functional Selectivity Relationship Studies on A-86929 Analogs and Small Aryl Fragments toward the Discovery of Biased Dopamine D1 Receptor Agonists. ACS Chem Neurosci 2022; 13:1818-1831. [PMID: 35658399 DOI: 10.1021/acschemneuro.2c00235] [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: 11/30/2022] Open
Abstract
Dopamine regulates normal functions such as movement, reinforcement learning, and cognition, and its dysfunction has been implicated in multiple psychiatric and neurological disorders. Dopamine acts through D1- (D1R and D5R) and D2-class (D2R, D3R, and D4R) receptors and activates both G protein- and β-arrestin-dependent signaling pathways. Current dopamine receptor-based therapies are used to ameliorate motor deficits in Parkinson's disease or as antipsychotic medications for schizophrenia. These drugs show efficacy for ameliorating only some symptoms caused by dopamine dysfunction and are plagued by debilitating side effects. Studies in primates and rodents have shown that shifting the balance of dopamine receptor signaling toward the arrestin pathway can be beneficial for inducing normal movement, while reducing motor side effects such as dyskinesias, and can be efficacious at enhancing cognitive function compared to balanced agonists. Several structure-activity relationship (SAR) studies have embarked on discovering β-arrestin-biased dopamine agonists, focused on D2 partial agonists, noncatechol D1 agonists, and mixed D1/D2R dopamine receptor agonists. Here, we describe an SAR study to identify novel D1R β-arrestin-biased ligands using A-86929, a high-affinity D1R catechol agonist, as a core scaffold to identify chemical motifs responsible for β-arrestin-biased activity at both D1 and D2Rs. Most of the A-86929 analogs screened were G protein-biased, but none of them were exclusively arrestin-biased. Additionally, various small-fragment molecular probes displayed weak bias toward the β-arrestin pathway. Continued in-depth SFSR (structure-functional selectivity relationship) studies informed by structure determination, molecular modeling, and mutagenesis studies will facilitate the discovery of potent and efficacious arrestin-biased dopamine receptor ligands.
Collapse
Affiliation(s)
- Haoxi Li
- Department of Chemistry, University of Florida, Gainesville, Florida 32610, United States
| | - Rosa Mirabel
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida 32610, United States
| | - Joseph Zimmerman
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida 32610, United States
| | - Ion Ghiviriga
- Department of Chemistry, University of Florida, Gainesville, Florida 32610, United States
| | - Darian K Phidd
- Department of Chemistry, University of Florida, Gainesville, Florida 32610, United States
| | - Nicole Horenstein
- Department of Chemistry, University of Florida, Gainesville, Florida 32610, United States
| | - Nikhil M Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
27
|
Gao AY, Link PA, Bakri SJ, Haak AJ. Dopamine Receptor Signaling Regulates Fibrotic Activation of Retinal Pigmented Epithelial Cells. Am J Physiol Cell Physiol 2022; 323:C116-C124. [PMID: 35544697 DOI: 10.1152/ajpcell.00468.2021] [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: 11/22/2022]
Abstract
Retinal pigmented epithelial (RPE) cells play an important role in retinal fibrotic diseases such as proliferative vitreoretinopathy (PVR). The purpose of this study was to elucidate the involvement of dopamine receptor signaling in regulating the fibrotic activation of RPE cells. Dopamine receptor expression, the effect of dopamine on fibrotic activity, and dopamine production were measured in the human RPE cell line ARPE-19. The fibrotic activation of RPE cells was evaluated in response to treatments with selective dopamine receptor agonists and antagonists by measuring gene expression, migration, proliferation, and fibronectin deposition. DRD2 and DRD5 are the dominant dopaminergic receptors expressed in ARPE-19 cells and TGFβ stimulates enhances autocrine release of dopamine which we show further exasperates fibrotic activation. Finally, treatment with D2 dopamine receptor antagonists or D5 dopamine receptor agonists inhibits profibrotic gene expression, migration, proliferation, and fibronectin deposition and thus may serve as effective mechanisms for treating retinal fibrosis including PVR.
Collapse
Affiliation(s)
- Ashley Y Gao
- Mayo Clinic, Department of Ophthalmology, Rochester, MN, United States
| | - Patrick A Link
- Mayo Clinic, Department of Physiology and Biomedical Engineering, Rochester, MN, United States
| | - Sophie J Bakri
- Mayo Clinic, Department of Ophthalmology, Rochester, MN, United States
| | - Andrew J Haak
- Mayo Clinic, Department of Physiology and Biomedical Engineering, Rochester, MN, United States
| |
Collapse
|
28
|
Jones-Tabah J, Mohammad H, Paulus EG, Clarke PBS, Hébert TE. The Signaling and Pharmacology of the Dopamine D1 Receptor. Front Cell Neurosci 2022; 15:806618. [PMID: 35110997 PMCID: PMC8801442 DOI: 10.3389/fncel.2021.806618] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
The dopamine D1 receptor (D1R) is a Gαs/olf-coupled GPCR that is expressed in the midbrain and forebrain, regulating motor behavior, reward, motivational states, and cognitive processes. Although the D1R was initially identified as a promising drug target almost 40 years ago, the development of clinically useful ligands has until recently been hampered by a lack of suitable candidate molecules. The emergence of new non-catechol D1R agonists, biased agonists, and allosteric modulators has renewed clinical interest in drugs targeting this receptor, specifically for the treatment of motor impairment in Parkinson's Disease, and cognitive impairment in neuropsychiatric disorders. To develop better therapeutics, advances in ligand chemistry must be matched by an expanded understanding of D1R signaling across cell populations in the brain, and in disease states. Depending on the brain region, the D1R couples primarily to either Gαs or Gαolf through which it activates a cAMP/PKA-dependent signaling cascade that can regulate neuronal excitability, stimulate gene expression, and facilitate synaptic plasticity. However, like many GPCRs, the D1R can signal through multiple downstream pathways, and specific signaling signatures may differ between cell types or be altered in disease. To guide development of improved D1R ligands, it is important to understand how signaling unfolds in specific target cells, and how this signaling affects circuit function and behavior. In this review, we provide a summary of D1R-directed signaling in various neuronal populations and describe how specific pathways have been linked to physiological and behavioral outcomes. In addition, we address the current state of D1R drug development, including the pharmacology of newly developed non-catecholamine ligands, and discuss the potential utility of D1R-agonists in Parkinson's Disease and cognitive impairment.
Collapse
|
29
|
Moore TL, Young DA, Killiany RJ, Fonseca KR, Volfson D, Gray DL, Balice-Gordon R, Kozak R. The Effects of a Novel Non-catechol Dopamine Partial Agonist on Working Memory in the Aged Rhesus Monkey. Front Aging Neurosci 2021; 13:757850. [PMID: 34899271 PMCID: PMC8662559 DOI: 10.3389/fnagi.2021.757850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/23/2021] [Indexed: 11/13/2022] Open
Abstract
Aged-related declines in cognition, especially working memory and executive function, begin in middle-age and these abilities are known to be mediated by the prefrontal cortex (PFC) and more specifically the dopamine (DA) system within the PFC. In both humans and monkeys, there is significant evidence that the PFC is the first cortical region to change with age and the PFC appears to be particularly vulnerable to age-related loss of dopamine (DA). Therefore, the DA system is a strong candidate for therapeutic intervention to slow or reverse age related declines in cognition. In the present study, we administered a novel selective, potent, non-catechol DA D1 R agonist PF-6294 (Pfizer, Inc.) to aged female rhesus monkeys and assessed their performance on two benchmark tasks of working memory - the Delayed Non-match to Sample Task (DNMS) and Delayed Recognition Span Task (DRST). The DNMS task was administered first with the standard 10 s delay and then with 5 min delays, with and without distractors. The DRST was administered each day with four trials with unique sequences and one trial of a repeated sequence to assess evidence learning and retention. Overall, there was no significant effect of drug on performance on any aspect of the DNMS task. In contrast, we demonstrated that a middle range dose of PF-6294 significantly increased memory span on the DRST on the first and last days of testing and by the last day of testing the increased memory span was driven by the performance on the repeated trials.
Collapse
Affiliation(s)
- Tara L Moore
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States.,Center for Systems Neuroscience, Boston University, Boston, MA, United States
| | - Damon A Young
- Internal Medicine Research Unit Pfizer Worldwide Research, Development and Medical Pfizer Inc., Cambridge, MA, United States
| | - Ronald J Killiany
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States.,Center for Systems Neuroscience, Boston University, Boston, MA, United States
| | - Kari R Fonseca
- Medicine Design, Pfizer Worldwide Research, Development and Medical Pfizer Inc., Cambridge, MA, United States
| | - Dmitri Volfson
- Internal Medicine Research Unit Pfizer Worldwide Research, Development and Medical Pfizer Inc., Cambridge, MA, United States
| | - David L Gray
- Internal Medicine Research Unit Pfizer Worldwide Research, Development and Medical Pfizer Inc., Cambridge, MA, United States
| | - Rita Balice-Gordon
- Internal Medicine Research Unit Pfizer Worldwide Research, Development and Medical Pfizer Inc., Cambridge, MA, United States
| | - Rouba Kozak
- Internal Medicine Research Unit Pfizer Worldwide Research, Development and Medical Pfizer Inc., Cambridge, MA, United States
| |
Collapse
|
30
|
Yang Y. Functional Selectivity of Dopamine D 1 Receptor Signaling: Retrospect and Prospect. Int J Mol Sci 2021; 22:ijms222111914. [PMID: 34769344 PMCID: PMC8584964 DOI: 10.3390/ijms222111914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/18/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
Research progress on dopamine D1 receptors indicates that signaling no longer is limited to G protein-dependent cyclic adenosine monophosphate phosphorylation but also includes G protein-independent β-arrestin-related mitogen-activated protein kinase activation, regulation of ion channels, phospholipase C activation, and possibly more. This review summarizes recent studies revealing the complexity of D1 signaling and its clinical implications, and suggests functional selectivity as a promising strategy for drug discovery to magnify the merit of D1 signaling. Functional selectivity/biased receptor signaling has become a major research front because of its potential to improve therapeutics through precise targeting. Retrospective pharmacological review indicated that many D1 ligands have some degree of mild functional selectivity, and novel compounds with extreme bias at D1 signaling were reported recently. Behavioral and neurophysiological studies inspired new methods to investigate functional selectivity and gave insight into the biased signaling of several drugs. Results from recent clinical trials also supported D1 functional selectivity signaling as a promising strategy for discovery and development of better therapeutics.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
31
|
Abi-Dargham A, Javitch JA, Slifstein M, Anticevic A, Calkins ME, Cho YT, Fonteneau C, Gil R, Girgis R, Gur RE, Gur RC, Grinband J, Kantrowitz J, Kohler C, Krystal J, Murray J, Ranganathan M, Santamauro N, Van Snellenberg J, Tamayo Z, Wolf D, TRANSCENDS Group
D’SouzaDeepakSrihariVinodGueorguievaRalitzaPatelPrashantForselius-BielenKimberleeLuJingButlerAudreyFramGeenaAfriyie-AgyemangYvetteSelloniAlexandriaCadavidLauraGomez-LunaSandraGuptaAartiRadhakrishnanRajivRashidAliAkerRyanAbrahimPhilishaBassir NiaAnahitaSurtiToralKegelesLawrence SCarlsonMarleneGoldbergTerryGangwischJamesBenedictErinneGovilPreetikaBrazisStephanieMayerMegande la GarrigueNathalieFallonNatalkaBaumvollTopazAbeykoonSameeraPerlmanGregBobchinKellyElliottMarkSchmidtLyndsayRushSagePortAllisonHeffernanZacLaneyNinaKantorJennaHohingThomas, Gray D, Lieberman J. Dopamine D1R Receptor Stimulation as a Mechanistic Pro-cognitive Target for Schizophrenia. Schizophr Bull 2021; 48:199-210. [PMID: 34423843 PMCID: PMC8781338 DOI: 10.1093/schbul/sbab095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Decades of research have highlighted the importance of optimal stimulation of cortical dopaminergic receptors, particularly the D1R receptor (D1R), for prefrontal-mediated cognition. This mechanism is particularly relevant to the cognitive deficits in schizophrenia, given the abnormalities in cortical dopamine (DA) neurotransmission and in the expression of D1R. Despite the critical need for D1R-based therapeutics, many factors have complicated their development and prevented this important therapeutic target from being adequately interrogated. Challenges include determination of the optimal level of D1R stimulation needed to improve cognitive performance, especially when D1R expression levels, affinity states, DA levels, and the resulting D1R occupancy by DA, are not clearly known in schizophrenia, and may display great interindividual and intraindividual variability related to cognitive states and other physiological variables. These directly affect the selection of the level of stimulation necessary to correct the underlying neurobiology. The optimal mechanism for stimulation is also unknown and could include partial or full agonism, biased agonism, or positive allosteric modulation. Furthermore, the development of D1R targeting drugs has been complicated by complexities in extrapolating from in vitro affinity determinations to in vivo use. Prior D1R-targeted drugs have been unsuccessful due to poor bioavailability, pharmacokinetics, and insufficient target engagement at tolerable doses. Newer drugs have recently become available, and these must be tested in the context of carefully designed paradigms that address methodological challenges. In this paper, we discuss how a better understanding of these challenges has shaped our proposed experimental design for testing a new D1R/D5R partial agonist, PF-06412562, renamed CVL-562.
Collapse
Affiliation(s)
- Anissa Abi-Dargham
- Department of Psychiatry, Stony Brook Renaissance School of Medicine, Stony Brook, NY, USA,Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA,Department of Psychiatry, Yale University, New Haven, CT, USA,Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Cerevel Therapeutics Research and Development, Boston, MA, USA,To whom correspondence should be addressed; Tel: +(631) 885-0814; e-mail:
| | - Jonathan A Javitch
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| | - Mark Slifstein
- Department of Psychiatry, Stony Brook Renaissance School of Medicine, Stony Brook, NY, USA
| | - Alan Anticevic
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Monica E Calkins
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Youngsun T Cho
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Clara Fonteneau
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Roberto Gil
- Department of Psychiatry, Stony Brook Renaissance School of Medicine, Stony Brook, NY, USA
| | - Ragy Girgis
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| | - Raquel E Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruben C Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jack Grinband
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| | - Joshua Kantrowitz
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| | - Christian Kohler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Krystal
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - John Murray
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | | | | | - Jared Van Snellenberg
- Department of Psychiatry, Stony Brook Renaissance School of Medicine, Stony Brook, NY, USA
| | - Zailyn Tamayo
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Daniel Wolf
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - David Gray
- Cerevel Therapeutics Research and Development, Boston, MA, USA
| | - Jeffrey Lieberman
- Department of Psychiatry, New York State Psychaitric Institute, Columbia University, New York, NY, USA
| |
Collapse
|
32
|
Sanchez JE, Kc GB, Franco J, Allen WJ, Garcia JD, Sirimulla S. BiasNet: A Model to Predict Ligand Bias Toward GPCR Signaling. J Chem Inf Model 2021; 61:4190-4199. [PMID: 34397210 DOI: 10.1021/acs.jcim.1c00317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Signaling bias is a feature of many G protein-coupled receptor (GPCR) targeting drugs with potential clinical implications. Whether it is therapeutically advantageous for a drug to be G protein biased or β-arrestin biased depends on the context of the signaling pathway. Here, we explored GPCR ligands that exhibit biased signaling to gain insights into scaffolds and pharmacophores that lead to bias. More specifically, we considered BiasDB, a database containing information about GPCR biased ligands, and focused our analysis on ligands which show either a G protein or β-arrestin bias. Five different machine learning models were trained on these ligands using 15 different sets of features. Molecular fragments which were important for training the models were analyzed. Two of these fragments (number of secondary amines and number of aromatic amines) were more prevalent in β-arrestin biased ligands. After training a random forest model on HierS scaffolds, we found five scaffolds, which demonstrated G protein or β-arrestin bias. We also conducted t-SNE clustering, observing correspondence between unsupervised and supervised machine learning methods. To increase the applicability of our work, we developed a web implementation of our models, which can predict bias based on user-provided SMILES, drug names, or PubChem CID. Our web implementation is available at: drugdiscovery.utep.edu/biasnet.
Collapse
Affiliation(s)
- Jason E Sanchez
- Computational Science Program, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Govinda B Kc
- Computational Science Program, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Julian Franco
- Mechanical Engineering, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - William J Allen
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, Texas 78758, United States
| | - Jesus David Garcia
- Computer Science, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Suman Sirimulla
- Computational Science Program, The University of Texas at El Paso, El Paso, Texas 79968, United States.,Computer Science, The University of Texas at El Paso, El Paso, Texas 79968, United States.,Department of Pharmaceutical Science, The University of Texas at El Paso, El Paso, Texas 79968, United States
| |
Collapse
|
33
|
Sun B, Feng D, Chu MLH, Fish I, Lovera S, Sands ZA, Kelm S, Valade A, Wood M, Ceska T, Kobilka TS, Lebon F, Kobilka BK. Crystal structure of dopamine D1 receptor in complex with G protein and a non-catechol agonist. Nat Commun 2021; 12:3305. [PMID: 34083522 PMCID: PMC8175458 DOI: 10.1038/s41467-021-23519-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 04/29/2021] [Indexed: 02/04/2023] Open
Abstract
Dopamine D1 receptor (D1R) is an important drug target implicated in many psychiatric and neurological disorders. Selective agonism of D1R are sought to be the therapeutic strategy for these disorders. Most selective D1R agonists share a dopamine-like catechol moiety in their molecular structure, and their therapeutic potential is therefore limited by poor pharmacological properties in vivo. Recently, a class of non-catechol D1R selective agonists with a distinct scaffold and pharmacological properties were reported. Here, we report the crystal structure of D1R in complex with stimulatory G protein (Gs) and a non-catechol agonist Compound 1 at 3.8 Å resolution. The structure reveals the ligand bound to D1R in an extended conformation, spanning from the orthosteric site to extracellular loop 2 (ECL2). Structural analysis reveals that the unique features of D1R ligand binding pocket explains the remarkable selectivity of this scaffold for D1R over other aminergic receptors, and sheds light on the mechanism for D1R activation by the non-catechol agonist.
Collapse
Affiliation(s)
| | - Dan Feng
- ConfometRx, Inc., Santa Clara, CA, USA
| | | | | | | | - Zara A Sands
- UCB Pharma, Braine-l'Alleud, Belgium
- Confo Therapeutics, Zwijnaarde, Belgium
| | | | | | | | | | | | | | - Brian K Kobilka
- ConfometRx, Inc., Santa Clara, CA, USA.
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
34
|
Guo M, Xiang T, Li M, Sun Y, Sun S, Chen D, Jia Q, Li Y, Yao X, Wang X, Zhang X, He F, Wang M. Effects of intrastriatal injection of the dopamine receptor agonist SKF38393 and quinpirole on locomotor behavior in hemiparkinsonism rats. Behav Brain Res 2021; 411:113339. [PMID: 33945831 DOI: 10.1016/j.bbr.2021.113339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 12/27/2022]
Abstract
Dopamine (DA) in the striatum is essential to influence motor behavior and may lead to movement impairment in Parkinson's disease (PD). The present study examined the different functions of the DA D1 receptor (D1R) and DA D2 receptor (D2R) by intrastriatal injection of the D1R agonist SKF38393 and the D2R agonist quinpirole in 6-hydroxydopamine (6-OHDA)-lesioned and control rats. All rats separately underwent dose-response behavior testing for SKF38393 (0, 0.5, 1.0, and 1.5 μg/site) or quinpirole (0, 1.0, 2.0, and 3.0 μg/site) to determine the effects of the optimal modulating threshold dose. Two behavior assessment indices, the time of latency to fall and the number of steps on a rotating treadmill, were used as reliable readouts of motor stimulation variables for quantifying the motor effects of the drugs. The findings indicate that at threshold doses, SKF38393 (1.0 μg/site) and quinpirole (1.0 μg/site) produce a dose-dependent increase in locomotor activity compared to vehicle injection. The ameliorated behavioral responses to either SKF38393 or quinpirole in lesioned rats were greater than those in unlesioned control rats. Moreover, the dose-dependent increase in locomotor capacity for quinpirole was greater than that for SKF38393 in lesioned rats. These results can clarify several key issues related to DA receptors directly and may provide a basis for exploring the potential of future selective dopamine therapies for PD in humans.
Collapse
Affiliation(s)
- Mengnan Guo
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China
| | - Tianyu Xiang
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China
| | - Min Li
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China
| | - Yue Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China
| | - Shuang Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China
| | - Dadian Chen
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China
| | - Qingmei Jia
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China
| | - Yuchuan Li
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China
| | - Xiaomeng Yao
- School of Nursing Qilu Institute of Technology, Jinan, 250200, People's Republic of China
| | - Xiaojun Wang
- The First Hospital Affiliated With Shandong First Medicine University, Jinan, People's Republic of China
| | - Xiao Zhang
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, 250200, People's Republic of China
| | - Feng He
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China.
| | - Min Wang
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, People's Republic of China.
| |
Collapse
|
35
|
Wilbraham D, Biglan KM, Svensson KA, Tsai M, Kielbasa W. Safety, Tolerability, and Pharmacokinetics of Mevidalen (LY3154207), a Centrally Acting Dopamine D1 Receptor-Positive Allosteric Modulator (D1PAM), in Healthy Subjects. Clin Pharmacol Drug Dev 2021; 10:393-403. [PMID: 33029934 PMCID: PMC8048550 DOI: 10.1002/cpdd.874] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/02/2020] [Indexed: 12/27/2022]
Abstract
Activation of the brain dopamine D1 receptor has attracted attention because of its promising role in neuropsychiatric diseases. Although efforts to develop D1 agonists have been challenging, a positive allosteric modulator (PAM), represents an attractive approach with potential better drug-like properties. Phase 1 single-ascending-dose (SAD; NCT03616795) and multiple-ascending-dose (MAD; NCT02562768) studies with the D1PAM mevidalen (LY3154207) were conducted with healthy subjects. There were no treatment-related serious adverse events (AEs) in these studies. In the SAD study, 25-200 mg administered orally showed dose-proportional pharmacokinetics (PK) and acute dose-related increases in systolic blood pressure (SBP) and diastolic blood pressure DBP) and pulse rate at doses ≥ 75 mg. AE related to central activation were seen at doses ≥ 75 mg. At 25 and 75 mg, central penetration of mevidalen was confirmed by measurement of mevidalen in cerebrospinal fluid. In the MAD study, once-daily doses of mevidalen at 15-150 mg for 14 days showed dose-proportional PK. Acute dose-dependent increases in SBP, DBP, and PR were observed on initial administration, but with repeated dosing the effects diminished and returned toward baseline levels. Overall, these findings support further investigation of mevidalen as a potential treatment for a range of neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | | | - Max Tsai
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | | |
Collapse
|
36
|
Harris SS, Urs NM. Targeting β-Arrestins in the Treatment of Psychiatric and Neurological Disorders. CNS Drugs 2021; 35:253-264. [PMID: 33651366 DOI: 10.1007/s40263-021-00796-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 12/23/2022]
Abstract
Therapies for psychiatric and neurological disorders have been in the development and refinement process for the past 5 decades. Yet, most of these therapies lack optimal therapeutic efficacy and have multiple debilitating side effects. Recent advances in understanding the pathophysiological processes of psychiatric and neurological disorders have revealed an important role for β-arrestins, which are important regulators of G-protein-coupled receptor (GPCR) function, including desensitization and intracellular signaling. These findings have pushed β-arrestins to the forefront as potential therapeutic targets. Here, we highlight current knowledge on β-arrestin functions in certain psychiatric and neurological disorders (schizophrenia, Parkinson's disease, and substance abuse disorders), and how this has been leveraged to develop new therapeutic strategies. Furthermore, we discuss the obstacles impacting the field of β-arrestin-based therapeutic development and future approaches that might help advance strategies to develop optimal β-arrestin-based therapies.
Collapse
Affiliation(s)
- Sharonda S Harris
- Department of Pharmacology and Therapeutics, University of Florida, 1200 Newell Dr, ARB-R5-140, Gainesville, FL, 32610, USA
| | - Nikhil M Urs
- Department of Pharmacology and Therapeutics, University of Florida, 1200 Newell Dr, ARB-R5-140, Gainesville, FL, 32610, USA.
| |
Collapse
|
37
|
Xiao P, Yan W, Gou L, Zhong YN, Kong L, Wu C, Wen X, Yuan Y, Cao S, Qu C, Yang X, Yang CC, Xia A, Hu Z, Zhang Q, He YH, Zhang DL, Zhang C, Hou GH, Liu H, Zhu L, Fu P, Yang S, Rosenbaum DM, Sun JP, Du Y, Zhang L, Yu X, Shao Z. Ligand recognition and allosteric regulation of DRD1-Gs signaling complexes. Cell 2021; 184:943-956.e18. [PMID: 33571432 PMCID: PMC11005940 DOI: 10.1016/j.cell.2021.01.028] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/01/2020] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Dopamine receptors, including D1- and D2-like receptors, are important therapeutic targets in a variety of neurological syndromes, as well as cardiovascular and kidney diseases. Here, we present five cryoelectron microscopy (cryo-EM) structures of the dopamine D1 receptor (DRD1) coupled to Gs heterotrimer in complex with three catechol-based agonists, a non-catechol agonist, and a positive allosteric modulator for endogenous dopamine. These structures revealed that a polar interaction network is essential for catecholamine-like agonist recognition, whereas specific motifs in the extended binding pocket were responsible for discriminating D1- from D2-like receptors. Moreover, allosteric binding at a distinct inner surface pocket improved the activity of DRD1 by stabilizing endogenous dopamine interaction at the orthosteric site. DRD1-Gs interface revealed key features that serve as determinants for G protein coupling. Together, our study provides a structural understanding of the ligand recognition, allosteric regulation, and G protein coupling mechanisms of DRD1.
Collapse
Affiliation(s)
- Peng Xiao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wei Yan
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Gou
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an 710049, China
| | - Ya-Ni Zhong
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Liangliang Kong
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Chao Wu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xin Wen
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yuan Yuan
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Sheng Cao
- School of Life and Health Sciences, Kobilka Institute of Innovative Drug Discovery, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Changxiu Qu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xin Yang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chuan-Cheng Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Anjie Xia
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenquan Hu
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Qianqian Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yong-Hao He
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Dao-Lai Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Chao Zhang
- Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Gui-Hua Hou
- Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Huanxiang Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Lizhe Zhu
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Ping Fu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shengyong Yang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Daniel M Rosenbaum
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Yang Du
- School of Life and Health Sciences, Kobilka Institute of Innovative Drug Discovery, Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China.
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
38
|
Mailman RB, Yang Y, Huang X. D 1, not D 2, dopamine receptor activation dramatically improves MPTP-induced parkinsonism unresponsive to levodopa. Eur J Pharmacol 2021; 892:173760. [PMID: 33279520 PMCID: PMC7861126 DOI: 10.1016/j.ejphar.2020.173760] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/15/2022]
Abstract
Levodopa is the standard-of-care for Parkinson's disease, but continued loss of dopamine neurons with disease progression decreases its bioconversion to dopamine, leading to increased side effects and decreased efficacy. In theory, dopamine agonists could equal levodopa, but no approved oral "dopamine agonist" matches the efficacy of levodopa. There are consistent data in both primate models and in Parkinson's disease showing that selective high intrinsic activity D1 agonists can equal levodopa in efficacy. There are, however, no data on whether such compounds would be effective in severe disease when levodopa efficacy is low or absent. We compared two approved antiparkinson drugs (levodopa and the D2/3 agonist bromocriptine) with the experimental selective D1 full agonist dihydrexidine in two severely parkinsonian MPTP-treated non-human primates. Bromocriptine caused no discernible improvement in parkinsonian signs, whereas levodopa caused a small transient improvement in one of the two subjects. Conversely, the full D1 agonist dihydrexidine caused a dramatic improvement in both subjects, decreasing parkinsonian signs by ca. 75%. No attenuation of dihydrexidine effects was observed when the two subjects were pretreated with the D2 antagonist remoxipride. These data provide evidence that selective D1 agonists may provide profound antiparkinson symptomatic relief even when the degree of nigrostriatal degeneration is so severe that current drugs are ineffective. Until effective disease-modifying therapies are discovered, high intrinsic activity D1 agonists may offer a major therapeutic advance in improving the quality of life, and potentially the longevity, of late stage Parkinson's patients.
Collapse
Affiliation(s)
- Richard B Mailman
- Departments of Pharmacology and NeurologyPenn State University College of Medicine Hershey PA 17033, USA.
| | - Yang Yang
- Departments of Pharmacology and NeurologyPenn State University College of Medicine Hershey PA 17033, USA.
| | - Xuemei Huang
- Departments of Pharmacology and NeurologyPenn State University College of Medicine Hershey PA 17033, USA.
| |
Collapse
|
39
|
Foley TL, Burchett W, Chen Q, Flanagan ME, Kapinos B, Li X, Montgomery JI, Ratnayake AS, Zhu H, Peakman MC. Selecting Approaches for Hit Identification and Increasing Options by Building the Efficient Discovery of Actionable Chemical Matter from DNA-Encoded Libraries. SLAS DISCOVERY 2021; 26:263-280. [PMID: 33412987 DOI: 10.1177/2472555220979589] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past 20 years, the toolbox for discovering small-molecule therapeutic starting points has expanded considerably. Pharmaceutical researchers can now choose from technologies that, in addition to traditional high-throughput knowledge-based and diversity screening, now include the screening of fragment and fragment-like libraries, affinity selection mass spectrometry, and selection against DNA-encoded libraries (DELs). Each of these techniques has its own unique combination of advantages and limitations that makes them more, or less, suitable for different target classes or discovery objectives, such as desired mechanism of action. Layered on top of this are the constraints of the drug-hunters themselves, including budgets, timelines, and available platform capacity; each of these can play a part in dictating the hit identification strategy for a discovery program. In this article, we discuss some of the factors that we use to govern our building of a hit identification roadmap for a program and describe the increasing role that DELs are playing in our discovery strategy. Furthermore, we share our learning during our initial exploration of DEL and highlight the approaches we have evolved to maximize the value returned from DEL selections. Topics addressed include the optimization of library design and production, reagent validation, data analysis, and hit confirmation. We describe how our thinking in these areas has led us to build a DEL platform that has begun to deliver tractable matter to our global discovery portfolio.
Collapse
Affiliation(s)
| | | | - Qiuxia Chen
- Lead Generation Unit, HitGen Inc., Chengdu, Shuangliu District, China
| | | | | | - Xianyang Li
- Lead Generation Unit, HitGen Inc., Chengdu, Shuangliu District, China
| | | | | | - Hongyao Zhu
- Simulation and Modelling Sciences, Pfizer Inc., Groton, CT, USA
| | | |
Collapse
|
40
|
Gurevich EV, Gurevich VV. GRKs as Modulators of Neurotransmitter Receptors. Cells 2020; 10:52. [PMID: 33396400 PMCID: PMC7823573 DOI: 10.3390/cells10010052] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 01/08/2023] Open
Abstract
Many receptors for neurotransmitters, such as dopamine, norepinephrine, acetylcholine, and neuropeptides, belong to the superfamily of G protein-coupled receptors (GPCRs). A general model posits that GPCRs undergo two-step homologous desensitization: the active receptor is phosphorylated by kinases of the G protein-coupled receptor kinase (GRK) family, whereupon arrestin proteins specifically bind active phosphorylated receptors, shutting down G protein-mediated signaling, facilitating receptor internalization, and initiating distinct signaling pathways via arrestin-based scaffolding. Here, we review the mechanisms of GRK-dependent regulation of neurotransmitter receptors, focusing on the diverse modes of GRK-mediated phosphorylation of receptor subtypes. The immediate signaling consequences of GRK-mediated receptor phosphorylation, such as arrestin recruitment, desensitization, and internalization/resensitization, are equally diverse, depending not only on the receptor subtype but also on phosphorylation by GRKs of select receptor residues. We discuss the signaling outcome as well as the biological and behavioral consequences of the GRK-dependent phosphorylation of neurotransmitter receptors where known.
Collapse
Affiliation(s)
- Eugenia V. Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 27232, USA;
| | | |
Collapse
|
41
|
Wang P, Felsing DE, Chen H, Stutz SJ, Murphy RE, Cunningham KA, Allen JA, Zhou J. Discovery of Potent and Brain-Penetrant GPR52 Agonist that Suppresses Psychostimulant Behavior. J Med Chem 2020; 63:13951-13972. [PMID: 33198466 DOI: 10.1021/acs.jmedchem.0c01498] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The G protein-coupled receptor 52 (GPR52) is an orphan receptor that is selectively expressed in the striatum and regulates various brain functions through activation of cAMP-dependent pathways. GPR52 has been identified as a promising therapeutic target for central nervous system disorders including schizophrenia and substance use disorders. Here, a series of novel GPR52 agonists were designed, synthesized, and evaluated based on compound 4. Several potent and efficacious GPR52 agonists (12c, 23a, 23d, 23e, 23f, and 23h) were identified with nanomolar range potency based on a systematic structure-activity relationship exploration. Further studies of 12c indicate enhanced efficacy, excellent target selectivity, and pharmacokinetic properties including good brain permeability. In vivo proof-of-concept investigations revealed that 12c displayed antipsychotic-like activity by significantly inhibiting amphetamine-induced hyperlocomotor behavior in mice. Collectively, our findings have resulted in an efficacious, brain-penetrant GPR52 agonist as a valuable pharmacological tool for investigating the physiological and therapeutic potential of GPR52 activation.
Collapse
Affiliation(s)
- Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Daniel E Felsing
- Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Sonja J Stutz
- Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Ryan E Murphy
- Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Kathryn A Cunningham
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States.,Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - John A Allen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States.,Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States.,Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
42
|
Cardiac dopamine D1 receptor triggers ventricular arrhythmia in chronic heart failure. Nat Commun 2020; 11:4364. [PMID: 32868781 PMCID: PMC7459304 DOI: 10.1038/s41467-020-18128-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/30/2020] [Indexed: 01/25/2023] Open
Abstract
Pathophysiological roles of cardiac dopamine system remain unknown. Here, we show the role of dopamine D1 receptor (D1R)-expressing cardiomyocytes (CMs) in triggering heart failure-associated ventricular arrhythmia. Comprehensive single-cell resolution analysis identifies the presence of D1R-expressing CMs in both heart failure model mice and in heart failure patients with sustained ventricular tachycardia. Overexpression of D1R in CMs disturbs normal calcium handling while CM-specific deletion of D1R ameliorates heart failure-associated ventricular arrhythmia. Thus, cardiac D1R has the potential to become a therapeutic target for preventing heart failure-associated ventricular arrhythmia.
Collapse
|
43
|
Martel JC, Gatti McArthur S. Dopamine Receptor Subtypes, Physiology and Pharmacology: New Ligands and Concepts in Schizophrenia. Front Pharmacol 2020; 11:1003. [PMID: 32765257 PMCID: PMC7379027 DOI: 10.3389/fphar.2020.01003] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Dopamine receptors are widely distributed within the brain where they play critical modulator roles on motor functions, motivation and drive, as well as cognition. The identification of five genes coding for different dopamine receptor subtypes, pharmacologically grouped as D1- (D1 and D5) or D2-like (D2S, D2L, D3, and D4) has allowed the demonstration of differential receptor function in specific neurocircuits. Recent observation on dopamine receptor signaling point at dopamine-glutamate-NMDA neurobiology as the most relevant in schizophrenia and for the development of new therapies. Progress in the chemistry of D1- and D2-like receptor ligands (agonists, antagonists, and partial agonists) has provided more selective compounds possibly able to target the dopamine receptors homo and heterodimers and address different schizophrenia symptoms. Moreover, an extensive evaluation of the functional effect of these agents on dopamine receptor coupling and intracellular signaling highlights important differences that could also result in highly differentiated clinical pharmacology. The review summarizes the recent advances in the field, addressing the relevance of emerging new targets in schizophrenia in particular in relation to the dopamine - glutamate NMDA systems interactions.
Collapse
|
44
|
Kozak R, Kiss T, Dlugolenski K, Johnson DE, Gorczyca RR, Kuszpit K, Harvey BD, Stolyar P, Sukoff Rizzo SJ, Hoffmann WE, Volfson D, Hajós M, Davoren JE, Abbott AL, Williams GV, Castner SA, Gray DL. Characterization of PF-6142, a Novel, Non-Catecholamine Dopamine Receptor D1 Agonist, in Murine and Nonhuman Primate Models of Dopaminergic Activation. Front Pharmacol 2020; 11:1005. [PMID: 32733245 PMCID: PMC7358525 DOI: 10.3389/fphar.2020.01005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022] Open
Abstract
Selective activation of dopamine D1 receptors remains a promising pro-cognitive therapeutic strategy awaiting robust clinical investigation. PF-6142 is a key example from a recently disclosed novel series of non-catechol agonists and partial agonists of the dopamine D1/5 receptors (D1R) that exhibit pharmacokinetic (PK) properties suitable for oral delivery. Given their reported potential for functionally biased signaling compared to known catechol-based selective agonists, and the promising rodent PK profile of PF-6142, we utilized relevant in vivo assays in male rodents and male and female non-human primates (NHP) to evaluate the pharmacology of this new series. Studies in rodents showed that PF-6142 increased locomotor activity and prefrontal cortex acetylcholine release, increased time spent in wakefulness, and desynchronized the EEG, like known D1R agonists. D1R selectivity of PF-6142 was supported by lack of effect in D1R knock-out mice and blocked response in the presence of the D1R antagonist SCH-23390. Further, PF-6142 improved performance in rodent models of NMDA receptor antagonist-induced cognitive dysfunction, such as MK-801-disrupted paired-pulse facilitation, and ketamine-disrupted working memory performance in the radial arm maze. Similarly, PF-6142 reversed ketamine-induced deficits in NHP performing the spatial delayed recognition task. Of importance, PF-6142 did not alter the efficacy of risperidone in assays predictive of antipsychotic-like effect in rodents including pre-pulse inhibition and conditioned avoidance responding. These data support the continued development of non-catechol based D1R agonists for the treatment of cognitive impairment associated with brain disorders including schizophrenia.
Collapse
Affiliation(s)
- Rouba Kozak
- Global Research and Development, Pfizer Inc., Groton, CT, United States
| | - Tamás Kiss
- Global Research and Development, Pfizer Inc., Groton, CT, United States
| | - Keith Dlugolenski
- Global Research and Development, Pfizer Inc., Groton, CT, United States
| | - David E Johnson
- Global Research and Development, Pfizer Inc., Groton, CT, United States
| | | | - Kyle Kuszpit
- Global Research and Development, Pfizer Inc., Groton, CT, United States
| | - Brian D Harvey
- Global Research and Development, Pfizer Inc., Groton, CT, United States
| | - Polina Stolyar
- Global Research and Development, Pfizer Inc., Groton, CT, United States
| | | | | | - Dmitri Volfson
- Global Research and Development, Pfizer Inc., Groton, CT, United States
| | - Mihaly Hajós
- Global Research and Development, Pfizer Inc., Groton, CT, United States.,Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, United States
| | | | - Amanda L Abbott
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Graham V Williams
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Stacy A Castner
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, United States
| | - David L Gray
- Global Research and Development, Pfizer Inc., Groton, CT, United States
| |
Collapse
|
45
|
Balice-Gordon R, Honey GD, Chatham C, Arce E, Duvvuri S, Naylor MG, Liu W, Xie Z, DeMartinis N, Harel BT, Braley GH, Kozak R, Park L, Gray DL. A Neurofunctional Domains Approach to Evaluate D1/D5 Dopamine Receptor Partial Agonism on Cognition and Motivation in Healthy Volunteers With Low Working Memory Capacity. Int J Neuropsychopharmacol 2020; 23:287-299. [PMID: 32055822 PMCID: PMC7251631 DOI: 10.1093/ijnp/pyaa007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/02/2020] [Accepted: 02/12/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Dopamine D1 receptor signaling plays key roles in core domains of neural function, including cognition and reward processing; however, many questions remain about the functions of circuits modulated by dopamine D1 receptor, largely because clinically viable, selective agonists have yet to be tested in humans. METHODS Using a novel, exploratory neurofunctional domains study design, we assessed the safety, tolerability, pharmacodynamics, and pharmacokinetics of PF-06412562, a selective D1/D5R partial agonist, in healthy male volunteers who met prespecified criteria for low working memory capacity. Functional magnetic resonance imaging, electrophysiologic endpoints, and behavioral paradigms were used to assess working memory, executive function, and motivation/reward processing following multiple-dose administration of PF-06412562. A total of 77 patients were assigned PF-06412562 (3 mg twice daily and 15 mg twice daily) or placebo administered for 5 to 7 days. Due to the exploratory nature of the study, it was neither powered for any specific treatment effect nor corrected for multiple comparisons. RESULTS Nominally significant improvements from baseline in cognitive endpoints were observed in all 3 groups; however, improvements in PF-06412562-treated patients were less than in placebo-treated participants. Motivation/reward processing endpoints were variable. PF-06412562 was safe and well tolerated, with no serious adverse events, severe adverse events, or adverse events leading to dose reduction or temporary discontinuation except for 1 permanent discontinuation due to increased orthostatic heart rate. CONCLUSIONS PF-06412562, in the dose range and patient population explored in this study, did not improve cognitive function or motivation/reward processing more than placebo over the 5- to 7-day treatment period. CLINICALTRIALS.GOV IDENTIFIER NCT02306876.
Collapse
Affiliation(s)
| | - Garry D Honey
- Pfizer Worldwide Research and Development, Cambridge, MA
| | | | - Estibaliz Arce
- Pfizer Worldwide Research and Development, Cambridge, MA
| | | | | | - Wenlei Liu
- Pfizer Worldwide Research and Development, Cambridge, MA
| | - Zhiyong Xie
- Pfizer Worldwide Research and Development, Cambridge, MA
| | | | - Brian T Harel
- Pfizer Worldwide Research and Development, Cambridge, MA
| | | | - Rouba Kozak
- Pfizer Worldwide Research and Development, Cambridge, MA
| | - Lovingly Park
- California Clinical Trials Medical Group/PAREXEL International, Glendale, CA
| | - David L Gray
- Pfizer Worldwide Research and Development, Cambridge, MA
| |
Collapse
|
46
|
Recent advances in dopaminergic strategies for the treatment of Parkinson's disease. Acta Pharmacol Sin 2020; 41:471-482. [PMID: 32112042 PMCID: PMC7471472 DOI: 10.1038/s41401-020-0365-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is the second most common progressive neurodegenerative disease worldwide. However, there is no available therapy reversing the neurodegenerative process of PD. Based on the loss of dopamine or dopaminergic dysfunction in PD patients, most of the current therapies focus on symptomatic relief to improve patient quality of life. As dopamine replacement treatment remains the most effective symptomatic pharmacotherapy for PD, herein we provide an overview of the current pharmacotherapies, summarize the clinical development status of novel dopaminergic agents, and highlight the challenge and opportunity of emerging preclinical dopaminergic approaches aimed at managing the features and progression of PD.
Collapse
|
47
|
Dopaminergic D 1 Receptor Stimulation Affects Effort and Risk Preferences. Biol Psychiatry 2020; 87:678-685. [PMID: 31668477 DOI: 10.1016/j.biopsych.2019.09.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Activation of D1 receptors has been related to successful goal-directed behavior, but it remains unclear whether D1 receptor activation causally tips the balance of weighing costs and benefits in humans. Here, we tested the impact of pharmacologically stimulated D1 receptors on sensitivity to risk, delay, and effort costs in economic choice and investigated whether D1 receptor stimulation would bias preferences toward options with increased costs in a cost-specific manner. METHODS In a randomized, double-blind, placebo-controlled, parallel-group phase 1 study, 120 healthy young volunteers received either placebo or 1 of 3 doses (6 mg, 15 mg, or 30 mg) of a novel, selective D1 agonist (PF-06412562). After drug administration, participants performed decision tasks measuring their preferences for risky, delayed, and effortful outcomes. RESULTS Higher doses of the D1 agonist increased the willingness to exert physical effort for reward as well as reduced the preference for risky outcomes. We observed no effects on preferences for delayed rewards. CONCLUSIONS The current results provide evidence that D1 receptor stimulation causally affects core aspects of cost-benefit decision making in humans.
Collapse
|
48
|
Soutschek A, Kozak R, de Martinis N, Howe W, Burke CJ, Fehr E, Jetter A, Tobler PN. Activation of D1 receptors affects human reactivity and flexibility to valued cues. Neuropsychopharmacology 2020; 45:780-785. [PMID: 31962344 PMCID: PMC7075935 DOI: 10.1038/s41386-020-0617-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/02/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022]
Abstract
Reward-predicting cues motivate goal-directed behavior, but in unstable environments humans must also be able to flexibly update cue-reward associations. While the capacity of reward cues to trigger motivation ('reactivity') as well as flexibility in cue-reward associations have been linked to the neurotransmitter dopamine in humans, the specific contribution of the dopamine D1 receptor family to these behaviors remained elusive. To fill this gap, we conducted a randomized, placebo-controlled, double-blind pharmacological study testing the impact of three different doses of a novel D1 agonist (relative to placebo) on reactivity to reward-predicting cues (Pavlovian-to-instrumental transfer) and flexibility of cue-outcome associations (reversal learning). We observed that the impact of the D1 agonist crucially depended on baseline working memory functioning, which has been identified as a proxy for baseline dopamine synthesis capacity. Specifically, increasing D1 receptor stimulation strengthened Pavlovian-to-instrumental transfer in individuals with high baseline working memory capacity. In contrast, higher doses of the D1 agonist improved reversal learning only in individuals with low baseline working memory functioning. Our findings suggest a crucial and baseline-dependent role of D1 receptor activation in controlling both cue reactivity and the flexibility of cue-reward associations.
Collapse
Affiliation(s)
- Alexander Soutschek
- Department of Psychology, Ludwig Maximilian University Munich, Munich, Germany. .,Zurich Center for Neuroeconomics, Department of Economics, University of Zurich, Zurich, Switzerland.
| | - Rouba Kozak
- 0000 0004 0447 7762grid.419849.9Takeda Pharmaceuticals International, Cambridge, MA USA
| | | | - William Howe
- 0000 0001 0694 4940grid.438526.eSchool of Neuroscience, Virginia Tech, Blacksburg, VA USA
| | - Christopher J. Burke
- 0000 0004 1937 0650grid.7400.3Zurich Center for Neuroeconomics, Department of Economics, University of Zurich, Zurich, Switzerland
| | - Ernst Fehr
- 0000 0004 1937 0650grid.7400.3Zurich Center for Neuroeconomics, Department of Economics, University of Zurich, Zurich, Switzerland ,Neuroscience Center Zurich, University of Zurich, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Alexander Jetter
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Philippe N. Tobler
- 0000 0004 1937 0650grid.7400.3Zurich Center for Neuroeconomics, Department of Economics, University of Zurich, Zurich, Switzerland ,Neuroscience Center Zurich, University of Zurich, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
49
|
Park H, Urs AN, Zimmerman J, Liu C, Wang Q, Urs NM. Structure-Functional-Selectivity Relationship Studies of Novel Apomorphine Analogs to Develop D1R/D2R Biased Ligands. ACS Med Chem Lett 2020; 11:385-392. [PMID: 32184974 DOI: 10.1021/acsmedchemlett.9b00575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/06/2020] [Indexed: 12/13/2022] Open
Abstract
Loss of dopamine neurons is central to the manifestation of Parkinson's disease motor symptoms. The dopamine precursor L-DOPA, the most commonly used therapeutic agent for Parkinson's disease, can restore normal movement yet cause side-effects such as dyskinesias upon prolonged administration. Dopamine D1 and D2 receptors activate G-protein- and arrestin-dependent signaling pathways that regulate various dopamine-dependent functions including locomotion. Studies have shown that shifting the balance of dopamine receptor signaling toward the arrestin pathway can be beneficial for inducing normal movement, while reducing dyskinesias. However, simultaneous activation of both D1 and D2Rs is required for robust locomotor activity. Thus, it is desirable to develop ligands targeting both D1 and D2Rs and their functional selectivity. Here, we report structure-functional-selectivity relationship (SFSR) studies of novel apomorphine analogs to identify structural motifs responsible for biased activity at both D1 and D2Rs.
Collapse
Affiliation(s)
- Hyejin Park
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Aarti N. Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville Florida 32610, United States
| | - Joseph Zimmerman
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville Florida 32610, United States
| | - Chuan Liu
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Qiu Wang
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Nikhil M. Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville Florida 32610, United States
| |
Collapse
|
50
|
Young D, Popiolek M, Trapa P, Fonseca KR, Brevard J, Gray DL, Kozak R. D1 Agonist Improved Movement of Parkinsonian Nonhuman Primates with Limited Dyskinesia Side Effects. ACS Chem Neurosci 2020; 11:560-566. [PMID: 31971364 DOI: 10.1021/acschemneuro.9b00589] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disease characterized by striatal dopaminergic loss. L-DOPA treatment replaces lost dopamine and enables motor function; however, eventually, fluctuating efficacy and side effects associated with its use become challenging for many patients. Here, we demonstrate, in a clinically translatable nonhuman primate model of parkinsonian motor symptoms, that treatment with the partial D1 receptor agonist CVL-751, formerly known as PF-06649751, is just as effective as L-DOPA in enabling movement and reducing disability. Importantly, CVL-751 efficacy is observed with less of the concomitant dyskinesia side effect associated with L-DOPA treatment. Data presented suggest that partial D1 agonists may be an effective and important treatment strategy for the management of Parkinson's patients.
Collapse
Affiliation(s)
| | | | | | | | - Julie Brevard
- Quanticare, Cambridge, Massachusetts 02139, United States
| | | | | |
Collapse
|