1
|
Bespalov A, Lütjens R, Doller D. Dusting off old blueprints: Is it time to reconsider metabotropic glutamate receptor 2 for therapeutic drug development? Pharmacol Biochem Behav 2025; 247:173908. [PMID: 39571688 DOI: 10.1016/j.pbb.2024.173908] [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: 10/09/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 01/28/2025]
Abstract
The metabotropic glutamate receptor 2 (mGlu2) is a heavily studied therapeutic target in neuropsychiatry for which we anticipate a renewed interest in the near future. We review the rationale and the outcome of clinical trials with mGlu2/3 receptor agonists in schizophrenia, a field of intense research since a seminal publication by Patel and colleagues (2007). We summarize evidence about selective, potent and safe agents with quantifiable CNS penetration that can be used to test hypotheses of mGlu2 receptors involvement in neuropsychiatric diseases. We summarize lessons learned from previous programs that should be considered to maximize the probability of success when targeting orthosteric and allosteric enhancement of mGlu2 receptor function in schizophrenia and beyond. First, we propose expanding our focus beyond presynaptic mGlu2 receptor stimulation in schizophrenia to novel hypotheses and that the choice of a therapeutic indication no longer be dictated by commercial opportunity but following science as a driver. Second, evidence on internal validity of preclinical studies supporting efficacy claims in the mGlu2 field is very limited. This gap will need to be closed when reviewing the rationale to re-initiate efforts in this field. Third, the pomaglumetad program was halted due to insufficient clinical efficacy, partly because of the inability to identify a treatment responder population. In preclinical studies, effects of mGlu2/3 receptor stimulation also seemed to vary significantly between laboratories. Definition of the responsive subject population and development of response-predicting biomarkers is therefore one of the main avenues of further research in the mGlu2 field.
Collapse
|
2
|
Jiang L, Liu N, Zhao F, Huang B, Kang D, Zhan P, Liu X. Discovery of GluN2A subtype-selective N-methyl-d-aspartate (NMDA) receptor ligands. Acta Pharm Sin B 2024; 14:1987-2005. [PMID: 38799621 PMCID: PMC11119548 DOI: 10.1016/j.apsb.2024.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/04/2023] [Accepted: 12/28/2023] [Indexed: 05/29/2024] Open
Abstract
The N-methyl-d-aspartate (NMDA) receptors, which belong to the ionotropic Glutamate receptors, constitute a family of ligand-gated ion channels. Within the various subtypes of NMDA receptors, the GluN1/2A subtype plays a significant role in central nervous system (CNS) disorders. The present article aims to provide a comprehensive review of ligands targeting GluN2A-containing NMDA receptors, encompassing negative allosteric modulators (NAMs), positive allosteric modulators (PAMs) and competitive antagonists. Moreover, the ligands' structure-activity relationships (SARs) and the binding models of representative ligands are also discussed, providing valuable insights for the clinical rational design of effective drugs targeting CNS diseases.
Collapse
Affiliation(s)
| | | | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Boshi Huang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
3
|
Qi X, Yu X, Wei L, Jiang H, Dong J, Li H, Wei Y, Zhao L, Deng W, Guo W, Hu X, Li T. Novel α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR) potentiator LT-102: A promising therapeutic agent for treating cognitive impairment associated with schizophrenia. CNS Neurosci Ther 2024; 30:e14713. [PMID: 38615362 PMCID: PMC11016348 DOI: 10.1111/cns.14713] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 03/07/2024] [Accepted: 03/23/2024] [Indexed: 04/16/2024] Open
Abstract
AIMS We aimed to evaluate the potential of a novel selective α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR) potentiator, LT-102, in treating cognitive impairments associated with schizophrenia (CIAS) and elucidating its mechanism of action. METHODS The activity of LT-102 was examined by Ca2+ influx assays and patch-clamp in rat primary hippocampal neurons. The structure of the complex was determined by X-ray crystallography. The selectivity of LT-102 was evaluated by hERG tail current recording and kinase-inhibition assays. The electrophysiological characterization of LT-102 was characterized by patch-clamp recording in mouse hippocampal slices. The expression and phosphorylation levels of proteins were examined by Western blotting. Cognitive function was assessed using the Morris water maze and novel object recognition tests. RESULTS LT-102 is a novel and selective AMPAR potentiator with little agonistic effect, which binds to the allosteric site formed by the intradimer interface of AMPAR's GluA2 subunit. Treatment with LT-102 facilitated long-term potentiation in mouse hippocampal slices and reversed cognitive deficits in a phencyclidine-induced mouse model. Additionally, LT-102 treatment increased the protein level of brain-derived neurotrophic factor and the phosphorylation of GluA1 in primary neurons and hippocampal tissues. CONCLUSION We conclude that LT-102 ameliorates cognitive impairments in a phencyclidine-induced model of schizophrenia by enhancing synaptic function, which could make it a potential therapeutic candidate for CIAS.
Collapse
Affiliation(s)
- Xueyu Qi
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Xueli Yu
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Long Wei
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Han Jiang
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Jiangwen Dong
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Hongxing Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Yingying Wei
- The Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Liansheng Zhao
- The Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Wei Deng
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Wanjun Guo
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Xun Hu
- The Clinical Research Center and Department of Pathology, The Second Affiliated HospitalZhejiang University School of MedicineZhejiangHangzhouChina
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| |
Collapse
|
4
|
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
|
5
|
Tyagi S, Mishra R, Mazumder R, Mazumder A. Current Market Potential and Prospects of Copper-based Pyridine Derivatives: A Review. Curr Mol Med 2024; 24:1111-1123. [PMID: 37496249 DOI: 10.2174/1566524023666230726160056] [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: 01/19/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 07/28/2023]
Abstract
Nicotine, minodronic acid, nicotinamide (niacin), zolpidem, zolimidine, and other pyridine-based chemicals play vital roles in medicine and biology. Pyridinecontaining drugs are widely available on the market to treat a wide range of human ailments. As a result of these advances, pyridine research is continually expanding, and there are now higher expectations for how it may aid in the treatment of numerous ailments. This evaluation incorporates data acquired from sources, like PubMed, to provide a thorough summary of the approved drugs and bioactivity data for compounds containing pyridine. Most of the reactions discussed in this article will provide readers with a deeper understanding of various pyridine-related examples, which is necessary for the creation of copper catalysis-based synthetic processes that are more accessible, secure, environmentally friendly, and practical, and that also have higher accuracy and selectivity. This paper also discusses significant innovations in the multi-component copper-catalyzed synthesis of N-heterocycles (pyridine), with the aim of developing precise, cost-effective, and environmentally friendly oxygenation and oxidation synthetic methods for the future synthesis of additional novel pyridine base analogs. Therefore, the review article will serve as a novel platform for researchers investigating copperbased pyridine compounds.
Collapse
Affiliation(s)
- Shivani Tyagi
- Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-2, Plot 19, Greater Noida, 201306, India
| | - Rakhi Mishra
- Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-2, Plot 19, Greater Noida, 201306, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-2, Plot 19, Greater Noida, 201306, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-2, Plot 19, Greater Noida, 201306, India
| |
Collapse
|
6
|
Abstract
Diseases associated with nicotine dependence in the form of habitual tobacco use are a major cause of premature death in the United States. The majority of tobacco smokers will relapse within the first month of attempted abstinence. Smoking cessation agents increase the likelihood that smokers can achieve long-term abstinence. Nevertheless, currently available smoking cessation agents have limited utility and fail to prevent relapse in the majority of smokers. Pharmacotherapy is therefore an effective strategy to aid smoking cessation efforts but considerable risk of relapse persists even when the most efficacious medications currently available are used. The past decade has seen major breakthroughs in our understanding of the molecular, cellular, and systems-level actions of nicotine in the brain that contribute to the development and maintenance of habitual tobacco use. In parallel, large-scale human genetics studies have revealed allelic variants that influence vulnerability to tobacco use disorder. These advances have revealed targets for the development of novel smoking cessation agents. Here, we summarize current efforts to develop smoking cessation therapeutics and highlight opportunities for future efforts.
Collapse
Affiliation(s)
- Dana Lengel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paul J. Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute (DDI), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
7
|
Domanegg K, Sommer WH, Meinhardt MW. Psychedelic Targeting of Metabotropic Glutamate Receptor 2 and Its Implications for the Treatment of Alcoholism. Cells 2023; 12:963. [PMID: 36980303 PMCID: PMC10047550 DOI: 10.3390/cells12060963] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Alcohol abuse is a leading risk factor for the public health burden worldwide. Approved pharmacotherapies have demonstrated limited effectiveness over the last few decades in treating alcohol use disorders (AUD). New therapeutic approaches are therefore urgently needed. Historical and recent clinical trials using psychedelics in conjunction with psychotherapy demonstrated encouraging results in reducing heavy drinking in AUD patients, with psilocybin being the most promising candidate. While psychedelics are known to induce changes in gene expression and neuroplasticity, we still lack crucial information about how this specifically counteracts the alterations that occur in neuronal circuits throughout the course of addiction. This review synthesizes well-established knowledge from addiction research about pathophysiological mechanisms related to the metabotropic glutamate receptor 2 (mGlu2), with findings and theories on how mGlu2 connects to the major signaling pathways induced by psychedelics via serotonin 2A receptors (2AR). We provide literature evidence that mGlu2 and 2AR are able to regulate each other's downstream signaling pathways, either through monovalent crosstalk or through the formation of a 2AR-mGlu2 heteromer, and highlight epigenetic mechanisms by which 2ARs can modulate mGlu2 expression. Lastly, we discuss how these pathways might be targeted therapeutically to restore mGlu2 function in AUD patients, thereby reducing the propensity to relapse.
Collapse
Affiliation(s)
- Kevin Domanegg
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 69115 Heidelberg, Germany
| | - Wolfgang H. Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 69115 Heidelberg, Germany
- Bethanien Hospital for Psychiatry, Psychosomatics, and Psychotherapy Greifswald, 17489 Greifswald, Germany
| | - Marcus W. Meinhardt
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 69115 Heidelberg, Germany
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 69115 Heidelberg, Germany
| |
Collapse
|
8
|
Martínez-Pinteño A, Rodríguez N, Olivares D, Madero S, Gómez M, Prohens L, García-Rizo C, Mas S, Morén C, Parellada E, Gassó P. Early treatment with JNJ-46356479, a mGluR2 modulator, improves behavioral and neuropathological deficits in a postnatal ketamine mouse model of schizophrenia. Biomed Pharmacother 2023; 158:114079. [PMID: 36521250 DOI: 10.1016/j.biopha.2022.114079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/21/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
Positive allosteric modulators of the metabotropic glutamate receptor 2 (mGluR2), such as JNJ-46356479 (JNJ), may mitigate the glutamate storm during the early stages of schizophrenia (SZ), which could be especially useful in the treatment of cognitive and negative symptoms. We evaluated the efficacy of early treatment with JNJ or clozapine (CLZ) in reversing behavioral and neuropathological deficits induced in a postnatal ketamine (KET) mouse model of SZ. Mice exposed to KET (30 mg/kg) on postnatal days (PND) 7, 9, and 11 received JNJ or CLZ (10 mg/kg) daily in the adolescent period (PND 35-60). Mice exposed to KET did not show the expected preference for a novel object or for social novelty, but they recovered this preference with JNJ treatment. Similarly, KET group did not show the expected dishabituation in the fifth trial, but mice treated with JNJ or CLZ recovered an interest in the novel animal. Neuronal immunoreactivity also differed between treatment groups with mice exposed to KET showing a reduction in parvalbumin positive cells in the prefrontal cortex and decreased c-Fos expression in the hippocampus, which was normalized with the pharmacological treatment. JNJ-46356479 treatment in early stages may help improve the cognitive and negative symptoms, as well as certain neuropathological deficits, and may even obtain a better response than CLZ treatment. This may have relevant clinical translational applications since early treatment with mGluR2 modulators that inhibit glutamate release at the onset of critical phases of SZ may prevent or slow down the clinical deterioration of the disease.
Collapse
Affiliation(s)
| | - N Rodríguez
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - D Olivares
- Dept. of Basic Clinical Practice, University of Barcelona, Spain
| | - S Madero
- Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain
| | - M Gómez
- Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - L Prohens
- Dept. of Basic Clinical Practice, University of Barcelona, Spain
| | - C García-Rizo
- Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - S Mas
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - C Morén
- Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain
| | - E Parellada
- Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain.
| | - P Gassó
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain.
| |
Collapse
|
9
|
Neurotoxic/Neuroprotective Effects of Clozapine and the Positive Allosteric Modulator of mGluR2 JNJ-46356479 in Human Neuroblastoma Cell Cultures. Int J Mol Sci 2023; 24:ijms24032054. [PMID: 36768378 PMCID: PMC9916793 DOI: 10.3390/ijms24032054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Current antipsychotics (APs) effectively control positive psychotic symptoms, mainly by blocking dopamine (DA) D2 receptors, but have little effect on negative and cognitive symptoms. Increased glutamate (GLU) release would trigger neurotoxicity, leading to apoptosis and synaptic pruning, which is involved in the pathophysiology of schizophrenia. New pharmacological strategies are being developed such as positive allosteric modulators (PAMs) of the metabotropic GLU receptor 2 (mGluR2) that inhibit the presynaptic release of GLU. We previously reported that treatment of adult mice with JNJ-46356479 (JNJ), a recently developed mGluR2 PAM, partially improved neuropathological deficits and schizophrenia-like behavior in a postnatal ketamine mouse model. In the present study, we evaluated, for the first time, the putative neuroprotective and antiapoptotic activity of JNJ in a human neuroblastoma cell line and compared it with the effect of clozapine (CLZ) as a clinical AP with the highest efficacy and with apparent utility in managing negative symptoms. Specifically, we measured changes in cell viability, caspase 3 activity and apoptosis, as well as in the expression of key genes involved in survival and cell death, produced by CLZ and JNJ alone and in combination with a high DA or GLU concentration as apoptosis inducers. Our results suggest that JNJ is not neurotoxic and attenuates apoptosis, particularly by decreasing the caspase 3 activation induced by DA and GLU, as well as increasing and decreasing the number of viable and apoptotic cells, respectively, only when cultures were exposed to GLU. Its effects seem to be less neurotoxic and more neuroprotective than those observed with CLZ. Moreover, JNJ partially normalized altered expression levels of glycolytic genes, which could act as a protective factor and be related to its putative neuroprotective effect. More studies are needed to define the mechanisms of action of this GLU modulator and its potential to become a novel therapeutic agent for schizophrenia.
Collapse
|
10
|
Li SH, Abd-Elrahman KS, Ferguson SS. Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases. Pharmacol Ther 2022; 239:108275. [DOI: 10.1016/j.pharmthera.2022.108275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 10/15/2022]
|
11
|
Yuan G, Dhaynaut M, Lan Y, Guehl NJ, Huynh D, Iyengar SM, Afshar S, Jain MK, Pickett JE, Kang HJ, Wang H, Moon SH, Ondrechen MJ, Wang C, Shoup TM, El Fakhri G, Normandin MD, Brownell AL. Synthesis and Characterization of 5-(2-Fluoro-4-[ 11C]methoxyphenyl)-2,2-dimethyl-3,4-dihydro-2 H-pyrano[2,3- b]pyridine-7-carboxamide as a PET Imaging Ligand for Metabotropic Glutamate Receptor 2. J Med Chem 2022; 65:2593-2609. [PMID: 35089713 PMCID: PMC9434702 DOI: 10.1021/acs.jmedchem.1c02004] [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: 11/30/2022]
Abstract
Metabotropic glutamate receptor 2 (mGluR2) is a therapeutic target for several neuropsychiatric disorders. An mGluR2 function in etiology could be unveiled by positron emission tomography (PET). In this regard, 5-(2-fluoro-4-[11C]methoxyphenyl)-2,2-dimethyl-3,4-dihydro-2H-pyrano[2,3-b]pyridine-7-carboxamide ([11C]13, [11C]mG2N001), a potent negative allosteric modulator (NAM), was developed to support this endeavor. [11C]13 was synthesized via the O-[11C]methylation of phenol 24 with a high molar activity of 212 ± 76 GBq/μmol (n = 5) and excellent radiochemical purity (>99%). PET imaging of [11C]13 in rats demonstrated its superior brain heterogeneity and reduced accumulation with pretreatment of mGluR2 NAMs, VU6001966 (9) and MNI-137 (26), the extent of which revealed a time-dependent drug effect of the blocking agents. In a nonhuman primate, [11C]13 selectively accumulated in mGluR2-rich regions and resulted in high-contrast brain images. Therefore, [11C]13 is a potential candidate for translational PET imaging of the mGluR2 function.
Collapse
Affiliation(s)
- Gengyang Yuan
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Yu Lan
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Nicolas J Guehl
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Dalena Huynh
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Suhasini M Iyengar
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Sepideh Afshar
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Manish Kumar Jain
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Julie E Pickett
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Hye Jin Kang
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Hao Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Sung-Hyun Moon
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Mary Jo Ondrechen
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Timothy M Shoup
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
12
|
Brown DG, Wobst HJ. A survey of the clinical pipeline in neuroscience. Bioorg Med Chem Lett 2022; 56:128482. [PMID: 34864194 DOI: 10.1016/j.bmcl.2021.128482] [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: 10/03/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/02/2022]
Abstract
Many new first-in-class drugs for neuroscience indications have been introduced in the past decade including new treatments for migraine, amyotrophic lateral sclerosis, depression, and multiple sclerosis. However, significant unmet patient needs remain in areas such as chronic pain, neurodegeneration, psychiatric diseases, and epilepsy. This review summarizes some of the advanced clinical compounds for these indications. Additionally, current opportunities and challenges that remain with respect to genetic validation, biomarkers, and translational models are discussed.
Collapse
Affiliation(s)
- Dean G Brown
- Jnana Therapeutics, 6 Tide St, MA 02210, United States.
| | - Heike J Wobst
- Jnana Therapeutics, 6 Tide St, MA 02210, United States
| |
Collapse
|
13
|
Wang P, Gao X, Zhang K, Pei Q, Xu X, Yan F, Dong J, Jing C. Exploring the binding mechanism of positive allosteric modulators in human metabotropic glutamate receptor 2 using molecular dynamics simulations. Phys Chem Chem Phys 2021; 23:24125-24139. [PMID: 34596645 DOI: 10.1039/d1cp02157e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Positive allosteric modulators (PAMs) of human metabotropic glutamate receptor 2 (hmGlu2) are well-known in the treatment of psychiatric disorders for their higher selectivity and lower tolerance risk. A variety of PAMs have been reported over the last decade and two compounds were in Phase II clinical trials for schizophrenia and anxiety. These trials were discontinued on account of the unsatisfactory therapeutic efficacy, but PAMs were explored as novel treatments for addiction and epilepsy. Thus, it is still important to explore novel hmGlu2 PAMs in the near future. Nowadays, the challenges in optimizing drug potency and improving scaffold diversity for PAMs are the noncomprehensive character analyses of multiple scaffolds; the exploration of the binding modes of PAMs in the allosteric binding site have been proposed to reduce this difficulty. However, there has been no comprehensive research about the binding profiles of PAMs in the hmGlu2 receptor. To address this issue, this work explores the binding characters of eight PAMs representing five chemical series by multiple computational methods. As a result, the shared binding modes of the eight studied PAMs interacting with 15 residues in the allosteric binding site were defined. In addition, the reduced hydrophobicity with low electronegativity of R1, increased hydrophobicity with low negative electron density of R2 and the electronegativity of the linker were identified as indicators that regulate the affinity of PAMs. This finding agrees well with the physicochemical properties of reported multiple series PAMs. This comprehensive work sheds additional light on the binding mechanism and physicochemical regularity underlining PAMs affinity and could be further utilized as a structural and energetic blueprint for discovering and assessing novel PAMs for hmGlu2.
Collapse
Affiliation(s)
- Panpan Wang
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China.
| | - Xiaonan Gao
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China.
| | - Ke Zhang
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China.
| | - Qinglan Pei
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China.
| | - Xiaobo Xu
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China.
| | - Fengmei Yan
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China.
| | - Jianghong Dong
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China.
| | - Chenxi Jing
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China.
| |
Collapse
|
14
|
Donthamsetti P, Konrad DB, Hetzler B, Fu Z, Trauner D, Isacoff EY. Selective Photoswitchable Allosteric Agonist of a G Protein-Coupled Receptor. J Am Chem Soc 2021; 143:8951-8956. [PMID: 34115935 PMCID: PMC8227462 DOI: 10.1021/jacs.1c02586] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Indexed: 01/03/2023]
Abstract
G protein-coupled receptors (GPCRs) are the most common targets of drug discovery. However, the similarity between related GPCRs combined with the complex spatiotemporal dynamics of receptor activation in vivo has hindered drug development. Photopharmacology offers the possibility of using light to control the location and timing of drug action by incorporating a photoisomerizable azobenzene into a GPCR ligand, enabling rapid and reversible switching between an inactive and active configuration. Recent advances in this area include (i) photoagonists and photoantagonists that directly control receptor activity but are nonselective because they bind conserved sites, and (ii) photoallosteric modulators that bind selectively to nonconserved sites but indirectly control receptor activity by modulating the response to endogenous ligand. In this study, we designed a photoswitchable allosteric agonist that targets a nonconserved allosteric site for selectivity and activates the receptor on its own to provide direct control. This work culminated in the development of aBINA, a photoswitchable allosteric agonist that selectively activates the Gi/o-coupled metabotropic glutamate receptor 2 (mGluR2). aBINA is the first example of a new class of precision drugs for GPCRs and other clinically important signaling proteins.
Collapse
Affiliation(s)
- Prashant Donthamsetti
- Department
of Molecular and Cell Biology, University
of California, Berkeley, California 94720, United States
| | - David B. Konrad
- Department
of Pharmacy, Ludwig-Maximilians-Universität
München, Butenandtstraße 5-13, 81377 Munich, Germany
| | - Belinda Hetzler
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Zhu Fu
- Department
of Molecular and Cell Biology, University
of California, Berkeley, California 94720, United States
| | - Dirk Trauner
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Ehud Y. Isacoff
- Department
of Molecular and Cell Biology, University
of California, Berkeley, California 94720, United States
- Helen
Wills Neuroscience Institute, University
of California, Berkeley, California 94720, United States
- Molecular
Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| |
Collapse
|
15
|
Parellada E, Gassó P. Glutamate and microglia activation as a driver of dendritic apoptosis: a core pathophysiological mechanism to understand schizophrenia. Transl Psychiatry 2021; 11:271. [PMID: 33958577 PMCID: PMC8102516 DOI: 10.1038/s41398-021-01385-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/09/2021] [Accepted: 04/20/2021] [Indexed: 02/03/2023] Open
Abstract
Schizophrenia disorder remains an unsolved puzzle. However, the integration of recent findings from genetics, molecular biology, neuroimaging, animal models and translational clinical research offers evidence that the synaptic overpruning hypothesis of schizophrenia needs to be reassessed. During a critical period of neurodevelopment and owing to an imbalance of excitatory glutamatergic pyramidal neurons and inhibitory GABAergic interneurons, a regionally-located glutamate storm might occur, triggering excessive dendritic pruning with the activation of local dendritic apoptosis machinery. The apoptotic loss of dendritic spines would be aggravated by microglia activation through a recently described signaling system from complement abnormalities and proteins of the MHC, thus implicating the immune system in schizophrenia. Overpruning of dendritic spines coupled with aberrant synaptic plasticity, an essential function for learning and memory, would lead to brain misconnections and synaptic inefficiency underlying the primary negative symptoms and cognitive deficits of schizophrenia. This driving hypothesis has relevant therapeutic implications, including the importance of pharmacological interventions during the prodromal phase or the transition to psychosis, targeting apoptosis, microglia cells or the glutamate storm. Future research on apoptosis and brain integrity should combine brain imaging, CSF biomarkers, animal models and cell biology.
Collapse
Affiliation(s)
- Eduard Parellada
- Barcelona Clínic Schizophrenia Unit (BCSU). Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Catalonia, Spain.
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.
| | - Patricia Gassó
- Barcelona Clínic Schizophrenia Unit (BCSU). Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Catalonia, Spain
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Catalonia, Spain
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
16
|
Martínez-Pinteño A, García-Cerro S, Mas S, Torres T, Boloc D, Rodríguez N, Lafuente A, Gassó P, Arnaiz JA, Parellada E. The positive allosteric modulator of the mGlu2 receptor JNJ-46356479 partially improves neuropathological deficits and schizophrenia-like behaviors in a postnatal ketamine mice model. J Psychiatr Res 2020; 126:8-18. [PMID: 32407891 DOI: 10.1016/j.jpsychires.2020.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/10/2020] [Accepted: 04/18/2020] [Indexed: 12/30/2022]
Abstract
Current antipsychotics have limited efficacy in controlling cognitive and negative symptoms of schizophrenia (SZ). Glutamatergic dysregulation has been implicated in the pathophysiology of SZ, based on the capacity of N-methyl-D-aspartate receptor (NMDAR) antagonists such as ketamine (KET) to induce SZ-like behaviors. This could be related to their putative neuropathological effect on gamma-aminobutyric (GABAergic) interneurons expressing parvalbumin (PV), which would lead to a hyperglutamatergic condition. Metabotropic glutamate receptor 2 (mGluR2) negatively modulates glutamate release and has been considered a potential clinical target for novel antipsychotics drugs. Our aim was to evaluate the efficacy of JNJ-46356479 (JNJ), a positive allosteric modulator (PAM) of the mGluR2, in reversing neuropathological and behavioral deficits induced in a postnatal KET mice model of SZ. These animals presented impaired spontaneous alternation in the Y-maze test, suggesting deficits in spatial working memory, and a decrease in social motivation and memory, assessed in both the Three-Chamber and the Five Trial Social Memory tests. Interestingly, JNJ treatment of adult mice partially reversed these deficits. Mice treated with KET also showed a reduction in PV+ in the mPFC and dentate gyrus together with an increase in c-Fos expression in this hippocampal area. Compared to the control group, mice treated with KET + JNJ showed a similar PV density and c-Fos activity pattern. Our results suggest that pharmacological treatment with a PAM of the mGluR2 such as JNJ could help improve cognitive and negative symptoms related to SZ.
Collapse
Affiliation(s)
| | - Susana García-Cerro
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Spain
| | - Sergi Mas
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Spain; The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Teresa Torres
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Spain
| | - Daniel Boloc
- Department of Medicine, University of Barcelona, Spain
| | - Natalia Rodríguez
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Spain
| | - Amalia Lafuente
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Spain; The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Patricia Gassó
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Spain; The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Joan Albert Arnaiz
- Department of Basic Clinical Practice, Unit of Pharmacology, University of Barcelona, Spain; The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Clinical Pharmacology Department, Hospital Clínic de Barcelona, Spain.
| | - Eduard Parellada
- The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Department of Medicine, University of Barcelona, Spain; Barcelona Clinic Schizophrenia Unit (BCSU), Institute of Neuroscience, Hospital Clinic of Barcelona, University of Barcelona, Spain.
| |
Collapse
|
17
|
Hellyer SD, Aggarwal S, Chen ANY, Leach K, Lapinsky DJ, Gregory KJ. Development of Clickable Photoaffinity Ligands for Metabotropic Glutamate Receptor 2 Based on Two Positive Allosteric Modulator Chemotypes. ACS Chem Neurosci 2020; 11:1597-1609. [PMID: 32396330 DOI: 10.1021/acschemneuro.0c00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The metabotropic glutamate receptor 2 (mGlu2) is a transmembrane-spanning class C G protein-coupled receptor that is an attractive therapeutic target for multiple psychiatric and neurological disorders. A key challenge has been deciphering the contribution of mGlu2 relative to other closely related mGlu receptors in mediating different physiological responses, which could be achieved through the utilization of subtype selective pharmacological tools. In this respect, allosteric modulators that recognize ligand-binding sites distinct from the endogenous neurotransmitter glutamate offer the promise of higher receptor-subtype selectivity. We hypothesized that mGlu2-selective positive allosteric modulators could be derivatized to generate bifunctional pharmacological tools. Here we developed clickable photoaffinity probes for mGlu2 based on two different positive allosteric modulator scaffolds that retained similar pharmacological activity to parent compounds. We demonstrate successful probe-dependent incorporation of a commercially available clickable fluorophore using bioorthogonal conjugation. Importantly, we also show the limitations of using these probes to assess in situ fluorescence of mGlu2 in intact cells where significant nonspecific membrane binding is evident.
Collapse
Affiliation(s)
- Shane D. Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Shaili Aggarwal
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Amy N. Y. Chen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - David J. Lapinsky
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
18
|
Tan Y, Xu Y, Cheng C, Zheng C, Zeng W, Wang J, Zhang X, Yang X, Wang J, Yang X, Nie S, Cao X. LY354740 Reduces Extracellular Glutamate Concentration, Inhibits Phosphorylation of Fyn/NMDARs, and Expression of PLK2/pS129 α-Synuclein in Mice Treated With Acute or Sub-Acute MPTP. Front Pharmacol 2020; 11:183. [PMID: 32180729 PMCID: PMC7059821 DOI: 10.3389/fphar.2020.00183] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
Glutamate overactivity in basal ganglia critically contributes to the exacerbation of dopaminergic neuron degeneration in Parkinson's disease (PD). Activation of group II metabotropic glutamate receptors (mGlu2/3 receptors), which can decrease excitatory glutamate neurotransmission, provides an opportunity to slow down the degeneration of the dopaminergic system. However, the roles of mGlu2/3 receptors in relation to PD pathology were partially recognized. By using mGlu2/3 receptors agonist (LY354740) and mGlu2/3 receptors antagonist (LY341495) in mice challenged with different cumulative doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), we demonstrated that systemic injection of LY354740 reduced the level of extracellular glutamate and the extent of nigro-striatal degeneration in both acute and sub-acute MPTP mice, while LY341495 amplified the lesions in sub-acute MPTP mice only. LY354740 treatment improved behavioral dysfunctions mainly in acute MPTP mice and LY341495 treatment seemed to aggravate motor deficits in sub-acute MPTP mice. In addition, ligands of mGlu2/3 receptors also influenced the total amount of glutamate and dopamine in brain tissue. Interestingly, compared with normal mice, MPTP-treated mice abnormally up-regulated the expression of polo-like kinase 2 (PLK2)/pS129 α-synuclein and phosphorylation of Fyn/N-methyl-D-aspartate receptor subunit 2A/2B (GluN2A/2B). Both acute and sub-acute MPTP mice treated with LY354740 dose-dependently reduced all the above abnormal expression. Compared with MPTP mice treated with vehicle, mice pretreated with LY341495 exhibited much higher expression of p-Fyn Tyr416/p-GluN2B Tyr1472 and PLK2/pS129 α-synuclein in sub-acute MPTP mice models. Thus, our current data indicated that mGlu2/3 receptors ligands could influence MPTP-induced toxicity, which supported a role for mGlu2/3 receptors in PD pathogenesis.
Collapse
Affiliation(s)
- Yang Tan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chi Cheng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Zheng
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Weiqi Zeng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jialing Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomei Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuke Nie
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Koszła O, Targowska-Duda KM, Kędzierska E, Kaczor AA. In Vitro and In Vivo Models for the Investigation of Potential Drugs Against Schizophrenia. Biomolecules 2020; 10:biom10010160. [PMID: 31963851 PMCID: PMC7022578 DOI: 10.3390/biom10010160] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia (SZ) is a complex psychiatric disorder characterized by positive, negative, and cognitive symptoms, and is not satisfactorily treated by current antipsychotics. Progress in understanding the basic pathomechanism of the disease has been hampered by the lack of appropriate models. In order to develop modern drugs against SZ, efficient methods to study them in in vitro and in vivo models of this disease are required. In this review a short presentation of current hypotheses and concepts of SZ is followed by a description of current progress in the field of SZ experimental models. A critical discussion of advantages and limitations of in vitro models and pharmacological, genetic, and neurodevelopmental in vivo models for positive, negative, and cognitive symptoms of the disease is provided. In particular, this review concerns the important issue of how cellular and animal systems can help to meet the challenges of modeling the disease, which fully manifests only in humans, as experimental studies of SZ in humans are limited. Next, it is emphasized that novel clinical candidates should be evaluated in animal models for treatment-resistant SZ. In conclusion, the plurality of available in vitro and in vivo models is a consequence of the complex nature of SZ, and there are extensive possibilities for their integration. Future development of more efficient antipsychotics reflecting the pleiotropy of symptoms in SZ requires the incorporation of various models into one uniting model of the multifactorial disorder and use of this model for the evaluation of new drugs.
Collapse
Affiliation(s)
- Oliwia Koszła
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland;
| | - Katarzyna M. Targowska-Duda
- Department of Biopharmacy, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland
| | - Ewa Kędzierska
- Department of Pharmacology and Pharmacodynamics, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland;
| | - Agnieszka A. Kaczor
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Faculty of Pharmacy, Medical University of Lublin, 4A Chodźki St., PL-20093 Lublin, Poland;
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
- Correspondence:
| |
Collapse
|
20
|
Pérez-Benito L, Llinas del Torrent C, Pardo L, Tresadern G. The computational modeling of allosteric modulation of metabotropic glutamate receptors. FROM STRUCTURE TO CLINICAL DEVELOPMENT: ALLOSTERIC MODULATION OF G PROTEIN-COUPLED RECEPTORS 2020; 88:1-33. [DOI: 10.1016/bs.apha.2020.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|