1
|
Lorente JS, Sokolov AV, Ferguson G, Schiöth HB, Hauser AS, Gloriam DE. GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov 2025:10.1038/s41573-025-01139-y. [PMID: 40033110 DOI: 10.1038/s41573-025-01139-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 03/05/2025]
Abstract
G protein-coupled receptors (GPCRs) form one of the largest drug target families, reflecting their involvement in numerous pathophysiological processes. In this Review, we analyse drug discovery trends for the GPCR superfamily, covering compounds, targets and indications that have reached regulatory approval or that are being investigated in clinical trials. We find that there are 516 approved drugs targeting GPCRs, making up 36% of all approved drugs. These drugs act on 121 GPCR targets, one-third of all non-sensory GPCRs. Furthermore, 337 agents targeting 133 GPCRs, including 30 novel targets, are being investigated in clinical trials. Notably, 165 of these agents are approved drugs being tested for additional indications and novel agents are increasingly allosteric modulators and biologics. Remarkably, diabetes and obesity drugs targeting GPCRs had sales of nearly US $30 billion in 2023 and the numbers of clinical trials for GPCR modulators in the metabolic diseases, oncology and immunology areas are increasing strongly. Finally, we highlight the potential of untapped target-disease associations and pathway-biased signalling. Overall, this Review provides an up-to-date reference for the drugged and potentially druggable GPCRome to inform future GPCR drug discovery and development.
Collapse
Affiliation(s)
- Javier Sánchez Lorente
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aleksandr V Sokolov
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Gavin Ferguson
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- ALPX S.A.S., Grenoble, France
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Alexander S Hauser
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David E Gloriam
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Ma Q, Li H, Song Z, Deng Z, Huang W, Liu Q. Fueling the fight against cancer: Exploring the impact of branched-chain amino acid catalyzation on cancer and cancer immune microenvironment. Metabolism 2024; 161:156016. [PMID: 39222743 DOI: 10.1016/j.metabol.2024.156016] [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: 05/22/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Metabolism of Branched-chain amino acids (BCAAs) is essential for the nutrient necessities in mammals. Catalytic enzymes serve to direct the whole-body BCAAs oxidation which involve in the development of various metabolic disorders. The reprogrammed metabolic elements are also responsible for malignant oncogenic processes, and favor the formation of distinctive immunosuppressive microenvironment surrounding different cancers. The impotent immune surveillance related to BCAAs dysfunction is a novel topic to investigate. Here we focus on the BCAA catalysts that contribute to metabolic changes and dysregulated immune reactions in cancer progression. We summarize the current knowledge of BCAA catalyzation, highlighting the interesting roles of BCAA metabolism in the treatment of cancers.
Collapse
Affiliation(s)
- Qianquan Ma
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, China
| | - Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province
| | - Zhihao Song
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province.
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province.
| |
Collapse
|
3
|
Mielecki D, Salińska E. Group III metabotropic glutamate receptors: guardians against excitotoxicity in ischemic brain injury, with implications for neonatal contexts. Pharmacol Rep 2024; 76:1199-1218. [PMID: 39298028 PMCID: PMC11582219 DOI: 10.1007/s43440-024-00651-z] [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: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024]
Abstract
The group III metabotropic glutamate receptors (mGluRs), comprising mGluR4, mGluR6, mGluR7, and mGluR8, offer neuroprotective potential in mitigating excitotoxicity during ischemic brain injury, particularly in neonatal contexts. They are G-protein coupled receptors that inhibit adenylyl cyclase and reduce neurotransmitter release, mainly located presynaptically and acting as autoreceptors. This review aims to examine the differential expression and function of group III mGluRs across various brain regions such as the cortex, hippocampus, and cerebellum, with a special focus on the neonatal stage of development. Glutamate excitotoxicity plays a crucial role in the pathophysiology of brain ischemia in neonates. While ionotropic glutamate receptors are traditional targets for neuroprotection, their direct inhibition often leads to severe side effects due to their critical roles in normal neurotransmission and synaptic plasticity. Group III mGluRs provide a more nuanced and potentially safer approach by modulating rather than blocking glutamatergic transmission. Their downstream signaling cascade results in the regulation of intracellular calcium levels, neuronal hyperpolarization, and reduced neurotransmitter release, effectively decreasing excitotoxic signaling without completely suppressing essential glutamatergic functions. Importantly, the neuroprotective effects of group III mGluRs extend beyond direct modulation of glutamate release influencing glial cell function, neuroinflammation, and oxidative stress, all of which contribute to secondary injury cascades in brain ischemia. This comprehensive analysis of group III mGluRs multifaceted neuroprotective potential provides valuable insights for developing novel therapeutic strategies to combat excitotoxicity in neonatal ischemic brain injury.
Collapse
Affiliation(s)
- Damian Mielecki
- Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, Warsaw, 02-106, Poland.
| | - Elżbieta Salińska
- Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, Warsaw, 02-106, Poland
| |
Collapse
|
4
|
Zhuang R, Yan Z, Gao Y, Nurmamat A, Zhang S, Xiu M, Zhou Y, Pang Y, Li D, Zhao L, Liu X, Han Y. Evolutionary and functional analysis of metabotropic glutamate receptors in lampreys. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:1861-1877. [PMID: 38951427 DOI: 10.1007/s10695-024-01374-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024]
Abstract
The metabotropic glutamate receptor (mGluR, GRM) family is involved in multiple signaling pathways and regulates neurotransmitter release. However, the evolutionary history, distribution, and function of the mGluRs family in lampreys have not been determined. Therefore, we identified the mGluRs gene family in the genome of Lethenteron reissneri, which has been conserved throughout vertebrate evolution. We confirmed that Lr-GRM3, Lr-GRM5, and Lr-GRM7 encode three types of mGluRs in lamprey. Additionally, we investigated the distribution of Lr-GRM3 within this species by qPCR and Western blotting. Furthermore, we conducted RNA sequencing to investigate the molecular function of Lr-GRM3 in lamprey. Our gene expression profile revealed that, similar to that in jawed vertebrates, Lr-GRM3 participates in multiple signal transduction pathways and influences synaptic excitability in lampreys. Moreover, it also affects intestinal motility and the inflammatory response in lampreys. This study not only enhances the understanding of mGluRs' gene evolution but also highlights the conservation of GRM3's role in signal transduction while expanding our knowledge of its functions specifically within lampreys. In summary, our experimental findings provide valuable insights for studying both the evolution and functionality of the mGluRs family.
Collapse
Affiliation(s)
- Ruyu Zhuang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Zihao Yan
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Yicheng Gao
- The First Clinical College of China Medical University, Shenyang, 110001, China
| | - Ayqeqan Nurmamat
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Shuyuan Zhang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Min Xiu
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Yuesi Zhou
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Ya Pang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Ding Li
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Liang Zhao
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
| | - Xin Liu
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Yinglun Han
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China.
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian, 116081, China.
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China.
| |
Collapse
|
5
|
Strauss A, Gonzalez-Hernandez AJ, Lee J, Abreu N, Selvakumar P, Salas-Estrada L, Kristt M, Arefin A, Huynh K, Marx DC, Gilliland K, Melancon BJ, Filizola M, Meyerson J, Levitz J. Structural basis of positive allosteric modulation of metabotropic glutamate receptor activation and internalization. Nat Commun 2024; 15:6498. [PMID: 39090128 PMCID: PMC11294631 DOI: 10.1038/s41467-024-50548-x] [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: 03/21/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024] Open
Abstract
The metabotropic glutamate receptors (mGluRs) are neuromodulatory family C G protein coupled receptors which assemble as dimers and allosterically couple extracellular ligand binding domains (LBDs) to transmembrane domains (TMDs) to drive intracellular signaling. Pharmacologically, mGluRs can be targeted at the LBDs by glutamate and synthetic orthosteric compounds or at the TMDs by allosteric modulators. Despite the potential of allosteric compounds as therapeutics, an understanding of the functional and structural basis of their effects is limited. Here we use multiple approaches to dissect the functional and structural effects of orthosteric versus allosteric ligands. We find, using electrophysiological and live cell imaging assays, that both agonists and positive allosteric modulators (PAMs) can drive activation and internalization of group II and III mGluRs. The effects of PAMs are pleiotropic, boosting the maximal response to orthosteric agonists and serving independently as internalization-biased agonists across mGluR subtypes. Motivated by this and intersubunit FRET analyses, we determine cryo-electron microscopy structures of mGluR3 in the presence of either an agonist or antagonist alone or in combination with a PAM. These structures reveal PAM-driven re-shaping of intra- and inter-subunit conformations and provide evidence for a rolling TMD dimer interface activation pathway that controls G protein and beta-arrestin coupling.
Collapse
Affiliation(s)
- Alexa Strauss
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
- Tri-Institutional Program in Chemical Biology, New York, NY, 10065, USA
| | | | - Joon Lee
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Nohely Abreu
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Purushotham Selvakumar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Leslie Salas-Estrada
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Melanie Kristt
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Anisul Arefin
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kevin Huynh
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Dagan C Marx
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kristen Gilliland
- Warren Center for Neuroscience Drug Discovery at Vanderbilt University, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bruce J Melancon
- Warren Center for Neuroscience Drug Discovery at Vanderbilt University, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Joel Meyerson
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA.
- Tri-Institutional Program in Chemical Biology, New York, NY, 10065, USA.
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
6
|
Kahvandi N, Ebrahimi Z, Sharifi M, Karimi SA, Shahidi S, Salehi I, Haddadi R, Sarihi A. S-3,4-DCPG, a potent orthosteric agonist for the mGlu8 receptor, facilitates extinction and inhibits the reinstatement of morphine-induced conditioned place preference in male rats. Pharmacol Biochem Behav 2024; 240:173772. [PMID: 38653345 DOI: 10.1016/j.pbb.2024.173772] [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: 12/10/2023] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
The limbic system, particularly the NAc, shows a high concentration of metabotropic glutamate receptors (mGluRs). Recent evidence suggests the significant involvement of mGluRs in mental disorders, including substance abuse and addiction. The objective of this study was to examine the involvement of mGlu8 receptors in the NAc in the mechanisms underlying the extinction and reinstatement of conditioned place preference (CPP) induced by morphine. Male Wistar rats underwent surgical implantation of bilateral cannulas in the NAc and were assessed in a CPP protocol. In study 1 at the same time as the extinction phase, the rats were given varying doses of S-3,4-DCPG (0.03, 0.3, and 3 μg/0.5 μl). In study 2, rats that had undergone CPP extinction were given S-3,4-DCPG (0.03, 0.3, and 3 μg/0.5 μl) five minutes prior to receiving a subthreshold dose of morphine (1 mg/kg) in order to reactivate the previously extinguished morphine response. The findings demonstrated that administering S-3,4-DCPG directly into the accumbens nucleus resulted in a decrease in the duration of the CPP extinction phase. Moreover, dose-dependent administration of S-3,4-DCPG into the NAc inhibited CPP reinstatement. The observations imply that microinjection of S-3,4-DCPG as a potent orthosteric agonist with high selectivity for the mGlu8 receptor into the NAc promotes the process of extinction while concurrently exerting inhibitory effects on the reinstatement of morphine-induced CPP. This effect may be associated with the modulation of glutamate engagement within the NAc and the plasticity of reward pathways at the synaptic level.
Collapse
Affiliation(s)
- Nazanin Kahvandi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Physiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zahra Ebrahimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Physiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Sharifi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Asaad Karimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasool Haddadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
7
|
Meadows SM, Palaguachi F, Jang MW, Licht-Murava A, Barnett D, Zimmer TS, Zhou C, McDonough SR, Orr AL, Orr AG. Hippocampal astrocytes induce sex-dimorphic effects on memory. Cell Rep 2024; 43:114278. [PMID: 38795347 PMCID: PMC11234507 DOI: 10.1016/j.celrep.2024.114278] [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: 10/27/2023] [Revised: 03/19/2024] [Accepted: 05/09/2024] [Indexed: 05/27/2024] Open
Abstract
Astrocytic receptors influence cognitive function and can promote behavioral deficits in disease. These effects may vary based on variables such as biological sex, but it is not known if the effects of astrocytic receptors are dependent on sex. We leveraged in vivo gene editing and chemogenetics to examine the roles of astrocytic receptors in spatial memory and other processes. We show that reductions in metabotropic glutamate receptor 3 (mGluR3), the main astrocytic glutamate receptor in adults, impair memory in females but enhance memory in males. Similarly, increases in astrocytic mGluR3 levels have sex-dependent effects and enhance memory in females. mGluR3 manipulations also alter spatial search strategies during recall in a sex-specific manner. In addition, acute chemogenetic stimulation of Gi/o-coupled or Gs-coupled receptors in hippocampal astrocytes induces bidirectional and sex-dimorphic effects on memory. Thus, astrocytes are sex-dependent modulators of cognitive function and may promote sex differences in aging and disease.
Collapse
Affiliation(s)
- Samantha M Meadows
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10021, USA
| | - Fernando Palaguachi
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Minwoo Wendy Jang
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Avital Licht-Murava
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Daniel Barnett
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10021, USA
| | - Till S Zimmer
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Constance Zhou
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | - Samantha R McDonough
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10021, USA
| | - Adam L Orr
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10021, USA
| | - Anna G Orr
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10021, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA.
| |
Collapse
|
8
|
Panarello S, González-Díez A, Berizzi AE, Malhaire F, Borràs-Tudurí R, Rovira X, Serra C, Prézeau L, Pin JP, Goudet C, Llebaria A, Gómez-Santacana X. Photoswitchable positive allosteric modulators of metabotropic glutamate receptor 4 to improve selectivity. iScience 2024; 27:110123. [PMID: 38966572 PMCID: PMC11223089 DOI: 10.1016/j.isci.2024.110123] [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: 11/29/2023] [Revised: 03/15/2024] [Accepted: 05/24/2024] [Indexed: 07/06/2024] Open
Abstract
Metabotropic glutamate receptors (mGlu) regulate multiple functions in the nervous systems and are involved in several neurological disorders. However, selectively targeting individual mGlu subtypes with spatiotemporal precision is still an unmet need. Photopharmacology can address this concern through the utilization of photoswitchable compounds such as optogluram, which is a positive allosteric modulator (PAM) of mGlu4 that enables the precise control of physiological responses using light but does not have an optimal selectivity profile. Optogluram analogs were developed to obtain photoswitchable PAMs of mGlu4 receptor with an improved selectivity. Among them, optogluram-2 emerged as a photoswitchable ligand for mGlu4 receptor with activity as both PAM and allosteric agonists. It presents a higher selectivity and offers improved photoswitching of mGlu4 activity. These improved properties make optogluram-2 an excellent candidate to study the role of mGlu4 with a high spatiotemporal precision in systems where mGlu4 can be co-expressed with other mGlu receptors.
Collapse
Affiliation(s)
- Silvia Panarello
- MCS, Institute for Advanced Chemistry of Catalonia – CSIC, Barcelona, Spain
- PhD Program in Organic Chemistry of the University of Barcelona, Barcelona, Spain
| | - Aleix González-Díez
- MCS, Institute for Advanced Chemistry of Catalonia – CSIC, Barcelona, Spain
- PhD Program in Organic Chemistry of the University of Barcelona, Barcelona, Spain
| | - Alice E. Berizzi
- Institut de Génomique Fonctionnelle, Université de Montpellier, UMR 5203 CNRS and U 1191 INSERM, Montpellier, France
| | - Fanny Malhaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, UMR 5203 CNRS and U 1191 INSERM, Montpellier, France
| | | | - Xavier Rovira
- MCS, Institute for Advanced Chemistry of Catalonia – CSIC, Barcelona, Spain
| | - Carme Serra
- MCS, Institute for Advanced Chemistry of Catalonia – CSIC, Barcelona, Spain
| | - Laurent Prézeau
- Institut de Génomique Fonctionnelle, Université de Montpellier, UMR 5203 CNRS and U 1191 INSERM, Montpellier, France
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, Université de Montpellier, UMR 5203 CNRS and U 1191 INSERM, Montpellier, France
| | - Cyril Goudet
- Institut de Génomique Fonctionnelle, Université de Montpellier, UMR 5203 CNRS and U 1191 INSERM, Montpellier, France
| | - Amadeu Llebaria
- MCS, Institute for Advanced Chemistry of Catalonia – CSIC, Barcelona, Spain
| | - Xavier Gómez-Santacana
- MCS, Institute for Advanced Chemistry of Catalonia – CSIC, Barcelona, Spain
- Institut de Génomique Fonctionnelle, Université de Montpellier, UMR 5203 CNRS and U 1191 INSERM, Montpellier, France
| |
Collapse
|
9
|
Ferranti AS, Luessen DJ, Niswender CM. Novel pharmacological targets for GABAergic dysfunction in ADHD. Neuropharmacology 2024; 249:109897. [PMID: 38462041 PMCID: PMC11843668 DOI: 10.1016/j.neuropharm.2024.109897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
Attention deficit/hyperactivity disorder (ADHD) is a neurodevelopment disorder that affects approximately 5% of the population. The disorder is characterized by impulsivity, hyperactivity, and deficits in attention and cognition, although symptoms vary across patients due to the heterogenous and polygenic nature of the disorder. Stimulant medications are the standard of care treatment for ADHD patients, and their effectiveness has led to the dopaminergic hypothesis of ADHD in which deficits in dopaminergic signaling, especially in cortical brain regions, mechanistically underly ADHD pathophysiology. Despite their effectiveness in many individuals, almost one-third of patients do not respond to stimulant treatments and the long-term negative side effects of these medications remain unclear. Emerging clinical evidence is beginning to highlight an important role of dysregulated excitatory/inhibitory (E/I) balance in ADHD. These deficits in E/I balance are related to functional abnormalities in glutamate and Gamma-Aminobutyric Acid (GABA) signaling in the brain, with increasing emphasis placed on GABAergic interneurons driving specific aspects of ADHD pathophysiology. Recent genome-wide association studies (GWAS) have also highlighted how genes associated with GABA function are mutated in human populations with ADHD, resulting in the generation of several new genetic mouse models of ADHD. This review will discuss how GABAergic dysfunction underlies ADHD pathophysiology, and how specific receptors/proteins related to GABAergic interneuron dysfunction may be pharmacologically targeted to treat ADHD in subpopulations with specific comorbidities and symptom domains. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Anthony S Ferranti
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
| | - Deborah J Luessen
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
10
|
Zhang M, Chen T, Lu X, Lan X, Chen Z, Lu S. G protein-coupled receptors (GPCRs): advances in structures, mechanisms, and drug discovery. Signal Transduct Target Ther 2024; 9:88. [PMID: 38594257 PMCID: PMC11004190 DOI: 10.1038/s41392-024-01803-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of human membrane proteins and an important class of drug targets, play a role in maintaining numerous physiological processes. Agonist or antagonist, orthosteric effects or allosteric effects, and biased signaling or balanced signaling, characterize the complexity of GPCR dynamic features. In this study, we first review the structural advancements, activation mechanisms, and functional diversity of GPCRs. We then focus on GPCR drug discovery by revealing the detailed drug-target interactions and the underlying mechanisms of orthosteric drugs approved by the US Food and Drug Administration in the past five years. Particularly, an up-to-date analysis is performed on available GPCR structures complexed with synthetic small-molecule allosteric modulators to elucidate key receptor-ligand interactions and allosteric mechanisms. Finally, we highlight how the widespread GPCR-druggable allosteric sites can guide structure- or mechanism-based drug design and propose prospects of designing bitopic ligands for the future therapeutic potential of targeting this receptor family.
Collapse
Affiliation(s)
- Mingyang Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ting Chen
- Department of Cardiology, Changzheng Hospital, Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Xun Lu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaobing Lan
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Ziqiang Chen
- Department of Orthopedics, Changhai Hospital, Affiliated to Naval Medical University, Shanghai, 200433, China.
| | - Shaoyong Lu
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
11
|
Huang L, Xiao W, Wang Y, Li J, Gong J, Tu E, Long L, Xiao B, Yan X, Wan L. Metabotropic glutamate receptors (mGluRs) in epileptogenesis: an update on abnormal mGluRs signaling and its therapeutic implications. Neural Regen Res 2024; 19:360-368. [PMID: 37488891 PMCID: PMC10503602 DOI: 10.4103/1673-5374.379018] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/07/2023] [Accepted: 05/22/2023] [Indexed: 07/26/2023] Open
Abstract
Epilepsy is a neurological disorder characterized by high morbidity, high recurrence, and drug resistance. Enhanced signaling through the excitatory neurotransmitter glutamate is intricately associated with epilepsy. Metabotropic glutamate receptors (mGluRs) are G protein-coupled receptors activated by glutamate and are key regulators of neuronal and synaptic plasticity. Dysregulated mGluR signaling has been associated with various neurological disorders, and numerous studies have shown a close relationship between mGluRs expression/activity and the development of epilepsy. In this review, we first introduce the three groups of mGluRs and their associated signaling pathways. Then, we detail how these receptors influence epilepsy by describing the signaling cascades triggered by their activation and their neuroprotective or detrimental roles in epileptogenesis. In addition, strategies for pharmacological manipulation of these receptors during the treatment of epilepsy in experimental studies is also summarized. We hope that this review will provide a foundation for future studies on the development of mGluR-targeted antiepileptic drugs.
Collapse
Affiliation(s)
- Leyi Huang
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Wenjie Xiao
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Yan Wang
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Juan Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jiaoe Gong
- Department of Neurology, Hunan Children’s Hospital, Changsha, Hunan Province, China
| | - Ewen Tu
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan Province, China
| | - Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Lily Wan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| |
Collapse
|
12
|
Płoska A, Siekierzycka A, Cieślik P, Dobrucki LW, Kalinowski L, Wierońska JM. The Impact of LY487379 or CDPPB on eNOS Expression in the Mouse Brain and the Effect of Joint Administration of Compounds with NO • Releasers on MK-801- or Scopolamine-Driven Cognitive Dysfunction in Mice. Molecules 2024; 29:627. [PMID: 38338372 PMCID: PMC10856750 DOI: 10.3390/molecules29030627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
The role of endothelial nitric oxide synthase (eNOS) in the regulation of a variety of biological processes is well established, and its dysfunction contributes to brain pathologies, including schizophrenia or Alzheimer's disease (AD). Positive allosteric modulators (PAMs) of metabotropic glutamate (mGlu) receptors were shown to be effective procognitive compounds, but little is known about their impact on eNOS expression and stability. Here, we investigated the influence of the acute and chronic administration of LY487379 or CDPPB (mGlu2 and mGlu5 PAMs), on eNOS expression in the mouse brain and the effect of the joint administration of the ligands with nitric oxide (NO) releasers, spermineNONOate or DETANONOate, in different combinations of doses, on MK-801- or scopolamine-induced amnesia in the novel object recognition (NOR) test. Our results indicate that both compounds provoked eNOS monomer formation, and CDPPB at a dose of 5 mg/kg exaggerated the effect of MK-801 or scopolamine. The coadministration of spermineNONOate or DETANONOate enhanced the antiamnesic effect of CDPPB or LY487379. The best activity was observed for ineffective or moderate dose combinations. The results indicate that treatment with mGluR2 and mGluR5 PAMs may be burdened with the risk of promoting eNOS uncoupling through the induction of dimer dissociation. Administration of the lowest possible doses of the compounds with NO• donors, which themselves have procognitive efficacy, may be proposed for the treatment of schizophrenia or AD.
Collapse
Affiliation(s)
- Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland; (A.P.); (A.S.); (L.W.D.)
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland; (A.P.); (A.S.); (L.W.D.)
| | - Paulina Cieślik
- Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland;
| | - Lawrence W. Dobrucki
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland; (A.P.); (A.S.); (L.W.D.)
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, Urbana, IL 61801, USA
- Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland; (A.P.); (A.S.); (L.W.D.)
- BioTechMed Center, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Narutowicza Steet, 80-223 Gdansk, Poland
| | - Joanna M. Wierońska
- Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland;
| |
Collapse
|
13
|
Strauss A, Gonzalez-Hernandez AJ, Lee J, Abreu N, Selvakumar P, Salas-Estrada L, Kristt M, Marx DC, Gilliland K, Melancon BJ, Filizola M, Meyerson J, Levitz J. Structural basis of allosteric modulation of metabotropic glutamate receptor activation and desensitization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.13.552748. [PMID: 37645747 PMCID: PMC10461995 DOI: 10.1101/2023.08.13.552748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The metabotropic glutamate receptors (mGluRs) are neuromodulatory family C G protein coupled receptors which assemble as dimers and allosterically couple extracellular ligand binding domains (LBDs) to transmembrane domains (TMDs) to drive intracellular signaling. Pharmacologically, mGluRs can be targeted either at the LBDs by glutamate and synthetic orthosteric compounds or at the TMDs by allosteric modulators. Despite the potential of allosteric TMD-targeting compounds as therapeutics, an understanding of the functional and structural basis of their effects on mGluRs is limited. Here we use a battery of approaches to dissect the distinct functional and structural effects of orthosteric versus allosteric ligands. We find using electrophysiological and live cell imaging assays that both agonists and positive allosteric modulators (PAMs) can drive activation and desensitization of mGluRs. The effects of PAMs are pleiotropic, including both the ability to boost the maximal response to orthosteric agonists and to serve independently as desensitization-biased agonists across mGluR subtypes. Conformational sensors reveal PAM-driven inter-subunit re-arrangements at both the LBD and TMD. Motivated by this, we determine cryo-electron microscopy structures of mGluR3 in the presence of either an agonist or antagonist alone or in combination with a PAM. These structures reveal PAM-driven re-shaping of intra- and inter-subunit conformations and provide evidence for a rolling TMD dimer interface activation pathway that controls G protein and beta-arrestin coupling. Highlights -Agonists and PAMs drive mGluR activation, desensitization, and endocytosis-PAMs are desensitization-biased and synergistic with agonists-Four combinatorial ligand conditions reveal an ensemble of full-length mGluR structures with novel interfaces-Activation and desensitization involve rolling TMD interfaces which are re-shaped by PAM.
Collapse
|
14
|
Muraleetharan A, Wang Y, Rowe MC, Gould A, Gregory KJ, Hellyer SD. Rigorous Characterization of Allosteric Modulation of the Human Metabotropic Glutamate Receptor 1 Reveals Probe- and Assay-Dependent Pharmacology. Mol Pharmacol 2023; 103:325-338. [PMID: 36921922 DOI: 10.1124/molpharm.122.000664] [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: 12/15/2022] [Revised: 01/26/2023] [Accepted: 02/21/2023] [Indexed: 03/17/2023] Open
Abstract
Allosteric modulation of metabotropic glutamate receptor subtype 1 (mGlu1) represents a viable therapeutic target for treating numerous central nervous system disorders. Although multiple chemically distinct mGlu1 positive (PAMs) and negative (NAMs) allosteric modulators have been identified, drug discovery paradigms have not included rigorous pharmacological analysis. In the present study, we hypothesized that existing mGlu1 allosteric modulators possess unappreciated probe-dependent or biased pharmacology. Using human embryonic kidney 293 (HEK293A) cells stably expressing human mGlu1, we screened mGlu1 PAMs and NAMs from divergent chemical scaffolds for modulation of different mGlu1 orthosteric agonists in intracellular calcium (iCa2+) mobilization and inositol monophosphate (IP1) accumulation assays. Operational models of agonism and allosterism were used to derive estimates for important pharmacological parameters such as affinity, efficacy, and cooperativity. Modulation of glutamate and quisqualate-mediated iCa2+ mobilization revealed probe dependence at the level of affinity and cooperativity for both mGlu1 PAMs and NAMs. We also identified the previously described mGlu5 selective NAM PF-06462894 as an mGlu1 NAM with a different pharmacological profile from other NAMs. Differential profiles were also observed when comparing ligand pharmacology between iCa2+ mobilization and IP1 accumulation. The PAMs Ro67-4853 and CPPHA displayed apparent negative cooperativity for modulation of quisqualate affinity, and the NAMs CPCCOEt and PF-06462894 had a marked reduction in cooperativity with quisqualate in IP1 accumulation and upon extended incubation in iCa2+ mobilization assays. These data highlight the importance of rigorous assessment of mGlu1 modulator pharmacology to inform future drug discovery programs for mGlu1 allosteric modulators. SIGNIFICANCE STATEMENT: Metabotropic glutamate receptor subtype 1 (mGlu1) positive and negative allosteric modulators have therapeutic potential in multiple central nervous system disorders. We show that chemically distinct modulators display differential pharmacology with different orthosteric ligands and across divergent signaling pathways at human mGlu1. Such complexities in allosteric ligand pharmacology should be considered in future mGlu1 allosteric drug discovery programs.
Collapse
Affiliation(s)
- Ashwin Muraleetharan
- Drug Discovery Biology (A.M., Y.W., M.C.R., A.G., K.J.G., S.D.H.) and ARC Centre for Cryo-Electron Microscopy of Membrane Proteins (K.J.G.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Yuyang Wang
- Drug Discovery Biology (A.M., Y.W., M.C.R., A.G., K.J.G., S.D.H.) and ARC Centre for Cryo-Electron Microscopy of Membrane Proteins (K.J.G.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Matthew C Rowe
- Drug Discovery Biology (A.M., Y.W., M.C.R., A.G., K.J.G., S.D.H.) and ARC Centre for Cryo-Electron Microscopy of Membrane Proteins (K.J.G.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Ashleigh Gould
- Drug Discovery Biology (A.M., Y.W., M.C.R., A.G., K.J.G., S.D.H.) and ARC Centre for Cryo-Electron Microscopy of Membrane Proteins (K.J.G.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Karen J Gregory
- Drug Discovery Biology (A.M., Y.W., M.C.R., A.G., K.J.G., S.D.H.) and ARC Centre for Cryo-Electron Microscopy of Membrane Proteins (K.J.G.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Shane D Hellyer
- Drug Discovery Biology (A.M., Y.W., M.C.R., A.G., K.J.G., S.D.H.) and ARC Centre for Cryo-Electron Microscopy of Membrane Proteins (K.J.G.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
15
|
Free RB, Nilson AN, Boldizsar NM, Doyle TB, Rodriguiz RM, Pogorelov VM, Machino M, Lee KH, Bertz JW, Xu J, Lim HD, Dulcey AE, Mach RH, Woods JH, Lane JR, Shi L, Marugan JJ, Wetsel WC, Sibley DR. Identification and Characterization of ML321: A Novel and Highly Selective D 2 Dopamine Receptor Antagonist with Efficacy in Animal Models That Predict Atypical Antipsychotic Activity. ACS Pharmacol Transl Sci 2023; 6:151-170. [PMID: 36654757 PMCID: PMC9841785 DOI: 10.1021/acsptsci.2c00202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Indexed: 12/31/2022]
Abstract
We have developed and characterized a novel D2R antagonist with exceptional GPCR selectivity - ML321. In functional profiling screens of 168 different GPCRs, ML321 showed little activity beyond potent inhibition of the D2R and to a lesser extent the D3R, demonstrating excellent receptor selectivity. The D2R selectivity of ML321 may be related to the fact that, unlike other monoaminergic ligands, ML321 lacks a positively charged amine group and adopts a unique binding pose within the orthosteric binding site of the D2R. PET imaging studies in non-human primates demonstrated that ML321 penetrates the CNS and occupies the D2R in a dose-dependent manner. Behavioral paradigms in rats demonstrate that ML321 can selectively antagonize a D2R-mediated response (hypothermia) while not affecting a D3R-mediated response (yawning) using the same dose of drug, thus indicating exceptional in vivo selectivity. We also investigated the effects of ML321 in animal models that are predictive of antipsychotic efficacy in humans. We found that ML321 attenuates both amphetamine- and phencyclidine-induced locomotor activity and restored pre-pulse inhibition (PPI) of acoustic startle in a dose-dependent manner. Surprisingly, using doses that were maximally effective in both the locomotor and PPI studies, ML321 was relatively ineffective in promoting catalepsy. Kinetic studies revealed that ML321 exhibits slow-on and fast-off receptor binding rates, similar to those observed with atypical antipsychotics with reduced extrapyramidal side effects. Taken together, these observations suggest that ML321, or a derivative thereof, may exhibit ″atypical″ antipsychotic activity in humans with significantly fewer side effects than observed with the currently FDA-approved D2R antagonists.
Collapse
Affiliation(s)
- R. Benjamin Free
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| | - Ashley N. Nilson
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| | - Noelia M. Boldizsar
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| | - Trevor B. Doyle
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| | - Ramona M. Rodriguiz
- Department
of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine
Analysis Core Facility, Duke University
Medical Center, 354 Sands Building, 303 Research Drive, Durham, North Carolina27710, United States
| | - Vladimir M. Pogorelov
- Department
of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine
Analysis Core Facility, Duke University
Medical Center, 354 Sands Building, 303 Research Drive, Durham, North Carolina27710, United States
| | - Mayako Machino
- Computational
Chemistry and Molecular Biophysics Section, Molecular Targets and
Medications Discovery Branch, National Institute on Drug Abuse, Intramural
Research Program, National Institutes of
Health, 333 Cassell Drive, Baltimore, Maryland21224, United
States
| | - Kuo Hao Lee
- Computational
Chemistry and Molecular Biophysics Section, Molecular Targets and
Medications Discovery Branch, National Institute on Drug Abuse, Intramural
Research Program, National Institutes of
Health, 333 Cassell Drive, Baltimore, Maryland21224, United
States
| | - Jeremiah W. Bertz
- Department
of Pharmacology, University of Michigan
Medical School, 1150 W. Medical Center Dr., Ann Arbor, Michigan48109, United States
| | - Jinbin Xu
- Division
of Radiological Sciences, Department of Radiology, Mallinckrodt Institute
of Radiology, Washington University School
of Medicine, St. Louis, Missouri63110, United States
| | - Herman D. Lim
- Drug Discovery
Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC3052, Australia
| | - Andrés E. Dulcey
- Division
of Pre-Clinical Innovation, National Center for Advancing Translational
Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland20850, United States
| | - Robert H. Mach
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania19104, United States
| | - James H. Woods
- Department
of Pharmacology, University of Michigan
Medical School, 1150 W. Medical Center Dr., Ann Arbor, Michigan48109, United States
| | - J Robert Lane
- Centre
of Membrane Proteins and Receptors, Universities
of Birmingham and Nottingham, NottinghamNG7 2UH, United Kingdom
| | - Lei Shi
- Computational
Chemistry and Molecular Biophysics Section, Molecular Targets and
Medications Discovery Branch, National Institute on Drug Abuse, Intramural
Research Program, National Institutes of
Health, 333 Cassell Drive, Baltimore, Maryland21224, United
States
| | - Juan J. Marugan
- Division
of Pre-Clinical Innovation, National Center for Advancing Translational
Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland20850, United States
| | - William C. Wetsel
- Department
of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine
Analysis Core Facility, Duke University
Medical Center, 354 Sands Building, 303 Research Drive, Durham, North Carolina27710, United States
- Departments
of Neurobiology and Cell Biology, Duke University
Medical Center, 354 Sands Building, 303 Research Drive, Durham, North Carolina27710, United States
| | - David R. Sibley
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, 35 Convent Drive, MSC-3723, Bethesda, Maryland20892, United States
| |
Collapse
|
16
|
Holter KM, Pierce BE, Gould RW. Metabotropic glutamate receptor function and regulation of sleep-wake cycles. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:93-175. [PMID: 36868636 PMCID: PMC10973983 DOI: 10.1016/bs.irn.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Metabotropic glutamate (mGlu) receptors are the most abundant family of G-protein coupled receptors and are widely expressed throughout the central nervous system (CNS). Alterations in glutamate homeostasis, including dysregulations in mGlu receptor function, have been indicated as key contributors to multiple CNS disorders. Fluctuations in mGlu receptor expression and function also occur across diurnal sleep-wake cycles. Sleep disturbances including insomnia are frequently comorbid with neuropsychiatric, neurodevelopmental, and neurodegenerative conditions. These often precede behavioral symptoms and/or correlate with symptom severity and relapse. Chronic sleep disturbances may also be a consequence of primary symptom progression and can exacerbate neurodegeneration in disorders including Alzheimer's disease (AD). Thus, there is a bidirectional relationship between sleep disturbances and CNS disorders; disrupted sleep may serve as both a cause and a consequence of the disorder. Importantly, comorbid sleep disturbances are rarely a direct target of primary pharmacological treatments for neuropsychiatric disorders even though improving sleep can positively impact other symptom clusters. This chapter details known roles of mGlu receptor subtypes in both sleep-wake regulation and CNS disorders focusing on schizophrenia, major depressive disorder, post-traumatic stress disorder, AD, and substance use disorder (cocaine and opioid). In this chapter, preclinical electrophysiological, genetic, and pharmacological studies are described, and, when possible, human genetic, imaging, and post-mortem studies are also discussed. In addition to reviewing the important relationships between sleep, mGlu receptors, and CNS disorders, this chapter highlights the development of selective mGlu receptor ligands that hold promise for improving both primary symptoms and sleep disturbances.
Collapse
Affiliation(s)
- Kimberly M Holter
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Bethany E Pierce
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Robert W Gould
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
17
|
Liu YJ, Li YL, Fang ZH, Liao HL, Zhang YY, Lin J, Liu F, Shen JF. NMDARs mediate peripheral and central sensitization contributing to chronic orofacial pain. Front Cell Neurosci 2022; 16:999509. [PMID: 36238833 PMCID: PMC9553029 DOI: 10.3389/fncel.2022.999509] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Peripheral and central sensitizations of the trigeminal nervous system are the main mechanisms to promote the development and maintenance of chronic orofacial pain characterized by allodynia, hyperalgesia, and ectopic pain after trigeminal nerve injury or inflammation. Although the pathomechanisms of chronic orofacial pain are complex and not well known, sufficient clinical and preclinical evidence supports the contribution of the N-methyl-D-aspartate receptors (NMDARs, a subclass of ionotropic glutamate receptors) to the trigeminal nociceptive signal processing pathway under various pathological conditions. NMDARs not only have been implicated as a potential mediator of pain-related neuroplasticity in the peripheral nervous system (PNS) but also mediate excitatory synaptic transmission and synaptic plasticity in the central nervous system (CNS). In this review, we focus on the pivotal roles and mechanisms of NMDARs in the trigeminal nervous system under orofacial neuropathic and inflammatory pain. In particular, we summarize the types, components, and distribution of NMDARs in the trigeminal nervous system. Besides, we discuss the regulatory roles of neuron-nonneuronal cell/neuron-neuron communication mediated by NMDARs in the peripheral mechanisms of chronic orofacial pain following neuropathic injury and inflammation. Furthermore, we review the functional roles and mechanisms of NMDARs in the ascending and descending circuits under orofacial neuropathic and inflammatory pain conditions, which contribute to the central sensitization. These findings are not only relevant to understanding the underlying mechanisms, but also shed new light on the targeted therapy of chronic orofacial pain.
Collapse
Affiliation(s)
- Ya-Jing Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue-Ling Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhong-Han Fang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hong-Lin Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiu Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie-Fei Shen Fei Liu
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie-Fei Shen Fei Liu
| |
Collapse
|