1
|
Li P, Lei W, Dong Y, Wang X, Ye X, Tian Y, Yang Y, Liu J, Li N, Niu X, Wang X, Tian Y, Xu L, Yang Y, Liu J. mGluR7: The new player protecting the central nervous system. Ageing Res Rev 2024; 102:102554. [PMID: 39454762 DOI: 10.1016/j.arr.2024.102554] [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: 08/30/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
Metabotropic glutamate receptor 7 (mGluR7) belongs to the family of type III mGluR receptor, playing an important part in the central nervous system (CNS) through response to neurotransmitter regulation, reduction of excitatory toxicity, and early neuronal development. Drugs targeting mGluR7 (mGluR7 agonists, antagonists, and allosteric modulators) may be among the most promising agents for the treatment of CNS disorders, such as psychiatric disorders, neurodegenerative diseases, and neurodevelopmental impairments, though these potential therapies are at early stages and the data are still limited. In this review, we summarized the structure and function of mGluR7 and discussed recent progress on mGluR7 agonists and antagonists. A deeper understanding of mGluR7 will contribute to uncovering the molecular mechanisms of neuroprotection and providing a theoretical basis for the formulation of therapeutic strategies.
Collapse
Affiliation(s)
- Pan Li
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Ophthalmology, Xi'an No.1 Hospital, Faculty of Life Sciences and Medicine, Northwest University, 30 Fenxiang Road, Xi'an 710002, China
| | - Wangrui Lei
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yushu Dong
- Department of Neurosurgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Xingyan Ye
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yaru Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Jie Liu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Ning Li
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xiaochen Niu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xin Wang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yifan Tian
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Lu Xu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| | - Jincheng Liu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
2
|
Li Y, Xiao Z, Mori W, Sun J, Yamasaki T, Rong J, Fujinaga M, Chen J, Kumata K, Zhao C, Zhang Y, Collier TL, Hu K, Xie L, Zhou X, Zhang W, Song Z, Gao Y, Sun Z, Zhang K, Patel JS, Ran C, Chaudhary A, Sheffler DJ, Cosford NDP, Zhang L, Zhai C, Haider A, Yuan H, Zhang MR, Liang SH. Radiosynthesis and preclinical evaluation of a carbon-11 labeled PET ligand for imaging metabotropic glutamate receptor 7. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:306-315. [PMID: 39583907 PMCID: PMC11578812 DOI: 10.62347/puai9230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/29/2024] [Indexed: 11/26/2024]
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is a G protein-coupled receptor that is preferentially found in the active zone of neurotransmitter release in the central nervous system (CNS). mGlu7 plays a vital role in memory, learning, and neuronal development, rendering it a potential target for treating epilepsy, depression, and anxiety. The development of noninvasive imaging ligands targeting mGlu7 could help elucidate the functional significance of mGlu7 and accelerate drug discovery for neurological and psychiatric disorders. In this report, a novel carbon-11 labeled positron emission tomography (PET) tracer designated [11C]18 (codenamed MG7-2109) was synthesized via 11C-methylation in 23% decay-corrected radiochemical yield (RCY). In vitro serum stability, serum protein binding, in vitro autoradiography and ex vivo biodistribution studies of [11C]18 were conducted. Preliminary PET imaging results revealed a homogeneous distribution of [11C]18 and rapid clearance in rodent brains. This study provides valuable insights into the development of mGlu7-targeted PET tracer based on an isoxazolo(5,4-c)pyridine scaffold.
Collapse
Affiliation(s)
- Yinlong Li
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Zhiwei Xiao
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Wakana Mori
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and TechnologyChiba 263-8555, Japan
| | - Jiyun Sun
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and TechnologyChiba 263-8555, Japan
| | - Jian Rong
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Masayuki Fujinaga
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and TechnologyChiba 263-8555, Japan
| | - Jiahui Chen
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Katsushi Kumata
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and TechnologyChiba 263-8555, Japan
| | - Chunyu Zhao
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and TechnologyChiba 263-8555, Japan
| | - Thomas L Collier
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and TechnologyChiba 263-8555, Japan
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and TechnologyChiba 263-8555, Japan
| | - Xin Zhou
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Wei Zhang
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Zhendong Song
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Yabiao Gao
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Zhenkun Sun
- Department of Pharmacology and Chemical Biology, Emory University School of MedicineAtlanta, GA 30322, USA
| | - Kuo Zhang
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Jimmy S Patel
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Department of Radiation Oncology, Winship Cancer Institute of Emory UniversityAtlanta, GA 30322, USA
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical SchoolBoston, MA 02114, USA
| | - Ahmad Chaudhary
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Douglas J Sheffler
- Cancer Molecular Therapeutics Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Prebys Medical Discovery InstituteLa Jolla, CA 92037, USA
| | - Nicholas DP Cosford
- Cancer Molecular Therapeutics Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Prebys Medical Discovery InstituteLa Jolla, CA 92037, USA
| | - Linqi Zhang
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Chuangyan Zhai
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Ahmed Haider
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of MedicineAtlanta, GA 30322, USA
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and TechnologyChiba 263-8555, Japan
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| |
Collapse
|
3
|
Dogra S, Aguayo C, Xiang Z, Putnam J, Smith J, Johnston C, Foster DJ, Lindsley CW, Niswender CM, Conn PJ. Activation of Metabotropic Glutamate Receptor 3 Modulates Thalamo-accumbal Transmission and Rescues Schizophrenia-Like Physiological and Behavioral Deficits. Biol Psychiatry 2024; 96:230-242. [PMID: 38061467 PMCID: PMC11150332 DOI: 10.1016/j.biopsych.2023.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Polymorphisms in the gene encoding for metabotropic glutamate receptor 3 (mGlu3) are associated with an increased likelihood of schizophrenia diagnosis and can predict improvements in negative symptoms following treatment with antipsychotics. However, the mechanisms by which mGlu3 can regulate brain circuits involved in schizophrenia pathophysiology are not clear. METHODS We employed selective pharmacological tools and a variety of approaches including whole-cell patch-clamp electrophysiology, slice optogenetics, and fiber photometry to investigate the effects of mGlu3 activation on phencyclidine (PCP)-induced impairments in thalamo-accumbal transmission and sociability deficits. A chemogenetic approach was used to evaluate the role of thalamo-accumbal transmission in PCP-induced sociability deficits. RESULTS We first established that PCP treatment augmented excitatory transmission onto dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) in the nucleus accumbens (NAc) and induced sociability deficits. Our studies revealed a selective increase in glutamatergic synaptic transmission from thalamic afferents to D1-MSNs in the NAc shell. Chemogenetic silencing of thalamo-accumbal inputs rescued PCP-induced sociability deficits. Pharmacological activation of mGlu3 normalized PCP-induced impairments in thalamo-accumbal transmission and sociability deficits. Mechanistic studies revealed that mGlu3 activation induced robust long-term depression at synapses from the thalamic projections onto D1-MSNs in the NAc shell. CONCLUSIONS These data demonstrate that activation of mGlu3 decreases thalamo-accumbal transmission and thereby rescues sociability deficits in mouse modeling schizophrenia-like symptoms. These findings provide novel insights into the NAc-specific mechanisms and suggest that agents modulating glutamatergic signaling in the NAc may provide a promising approach for treating negative symptoms in schizophrenia.
Collapse
Affiliation(s)
- Shalini Dogra
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee.
| | - Caleb Aguayo
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee
| | - Zixiu Xiang
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee
| | - Jason Putnam
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee
| | - Joshua Smith
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee
| | - Curran Johnston
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Daniel J Foster
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee; Department of Chemistry, Vanderbilt University, Nashville, Tennessee; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, Tennessee
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University, Nashville, Tennessee; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, Tennessee
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee; Vanderbilt Kennedy Center, Vanderbilt University, Nashville, Tennessee; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
4
|
Zaki-Dizaji M, Abazari MF, Razzaghi H, Shkolnikov I, Christie BR. GRM7 deficiency, from excitotoxicity and neuroinflammation to neurodegeneration: Systematic review of GRM7 deficient patients. Brain Behav Immun Health 2024; 39:100808. [PMID: 38983774 PMCID: PMC11231722 DOI: 10.1016/j.bbih.2024.100808] [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: 12/20/2023] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
The metabotropic glutamate receptor 7 (mGluR7) is a presynaptic G-protein-coupled glutamate receptor that modulates neurotransmitter release and synaptic plasticity at presynaptic terminals. It is encoded by GRM7, and recently variants have been identified in patients with autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), developmental delay (DD), intellectual disability (ID), and brain malformations. To gain updated insights into the function of GRM7 and the phenotypic spectrum of genetic variations within this gene, we conducted a systematic review of relevant literature utilizing PubMed, Web of Science, and Scopus databases. Among the 14 articles meeting the inclusion criteria, a total of 42 patients (from 28 families) harboring confirmed mutations in the GRM7 gene have been documented. Specifically, there were 17 patients with heterozygous mutations, 20 patients with homozygous mutations, and 5 patients with compound heterozygous mutations. Common clinical features included intellectual behavioral disability, seizure/epilepsy, microcephaly, developmental delay, peripheral hypertonia and hypomyelination. Genotype-phenotype correlation was not clear and each variant had unique characteristics including gene dosage, mutant protein surface expression, and degradation pathway that result with a spectrum of phenotype manifestations through ASD or ADHD to severe DD/ID with brain malformations. Neuroinflammation may play a role in the development and/or progression of GRM7-related neurodegeneration along with excitotoxicity. The clinical and functional data presented here demonstrate that both autosomal dominant and recessive inheritance of GRM7 mutation can cause disease spectrum phenotypes through ASD or ADHD to severe DD/ID and seizure with brain malformations.
Collapse
Affiliation(s)
- Majid Zaki-Dizaji
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Foad Abazari
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
- Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada
| | - Hossein Razzaghi
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Irene Shkolnikov
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada
| |
Collapse
|
5
|
Dong HW, Weiss K, Baugh K, Meadows MJ, Niswender CM, Neul JL. Potentiation of the muscarinic acetylcholine receptor 1 modulates neurophysiological features in a mouse model of Rett syndrome. Neurotherapeutics 2024; 21:e00384. [PMID: 38880672 PMCID: PMC11284553 DOI: 10.1016/j.neurot.2024.e00384] [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: 04/17/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder primarily caused by mutations in the X chromosome-linked gene Methyl-CpG Binding Protein 2 (MECP2). Restoring MeCP2 expression after disease onset in a mouse model of RTT reverses phenotypes, providing hope for development of treatments for RTT. Translatable biomarkers of improvement and treatment responses have the potential to accelerate both preclinical and clinical evaluation of targeted therapies in RTT. Studies in people with and mouse models of RTT have identified neurophysiological features, such as auditory event-related potentials, that correlate with disease severity, suggesting that they could be useful as biomarkers of disease improvement or early treatment response. We recently demonstrated that treatment of RTT mice with a positive allosteric modulator (PAM) of muscarinic acetylcholine subtype 1 receptor (M1) improved phenotypes, suggesting that modulation of M1 activity is a potential therapy in RTT. To evaluate whether neurophysiological features could be useful biomarkers to assess the effects of M1 PAM treatment, we acutely administered the M1 PAM VU0486846 (VU846) at doses of 1, 3, 10 and 30 mg/kg in wildtype and RTT mice. This resulted in an inverted U-shaped dose response with maximal improvement of AEP features at 3 mg/kg but with no marked effect on basal EEG power or epileptiform discharges in RTT mice and no significant changes in wildtype mice. These findings suggest that M1 potentiation can improve neural circuit synchrony to auditory stimuli in RTT mice and that neurophysiological features have potential as pharmacodynamic or treatment-responsive biomarkers for preclinical and clinical evaluation of putative therapies in RTT.
Collapse
Affiliation(s)
- Hong-Wei Dong
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, USA; Vanderbilt University Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kelly Weiss
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Warren Center for Neuroscience Drug Discovery, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Kathryn Baugh
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, USA
| | - Mac J Meadows
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Warren Center for Neuroscience Drug Discovery, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Colleen M Niswender
- Vanderbilt University Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Warren Center for Neuroscience Drug Discovery, School of Medicine, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute for Chemical Biology, Nashville, TN, USA; Vanderbilt Brain Institute, Nashville, TN, USA.
| | - Jeffrey L Neul
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, USA; Vanderbilt University Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Nashville, TN, USA.
| |
Collapse
|
6
|
Illescas S, Diaz-Osorio Y, Serradell A, Toro-Soria L, Musokhranova U, Juliá-Palacios N, Ribeiro-Constante J, Altafaj X, Olivella M, O'Callaghan M, Darling A, Armstrong J, Artuch R, García-Cazorla À, Oyarzábal A. Metabolic characterization of neurogenetic disorders involving glutamatergic neurotransmission. J Inherit Metab Dis 2024; 47:551-569. [PMID: 37932875 DOI: 10.1002/jimd.12689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/28/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023]
Abstract
The study of inborn errors of neurotransmission has been mostly focused on monoamine disorders, GABAergic and glycinergic defects. The study of the glutamatergic synapse using the same approach than classic neurotransmitter disorders is challenging due to the lack of biomarkers in the CSF. A metabolomic approach can provide both insight into their molecular basis and outline novel therapeutic alternatives. We have performed a semi-targeted metabolomic analysis on CSF samples from 25 patients with neurogenetic disorders with an important expression in the glutamatergic synapse and 5 controls. Samples from patients diagnosed with MCP2, CDKL5-, GRINpathies and STXBP1-related encephalopathies were included. We have performed univariate (UVA) and multivariate statistical analysis (MVA), using Wilcoxon rank-sum test, principal component analysis (PCA), and OPLS-DA. By using the results of both analyses, we have identified the metabolites that were significantly altered and that were important in clustering the respective groups. On these, we performed pathway- and network-based analyses to define which metabolic pathways were possibly altered in each pathology. We have observed alterations in the tryptophan and branched-chain amino acid metabolism pathways, which interestingly converge on LAT1 transporter-dependency to cross the blood-brain barrier (BBB). Analysis of the expression of LAT1 transporter in brain samples from a mouse model of Rett syndrome (MECP2) revealed a decrease in the transporter expression, that was already noticeable at pre-symptomatic stages. The study of the glutamatergic synapse from this perspective advances the understanding of their pathophysiology, shining light on an understudied feature as is their metabolic signature.
Collapse
Affiliation(s)
- Sofía Illescas
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
| | - Yaiza Diaz-Osorio
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
| | - Anna Serradell
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
| | - Lucía Toro-Soria
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
| | - Uliana Musokhranova
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
| | - Natalia Juliá-Palacios
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
- Neurometabolic Unit, Hospital Sant Joan de Déu, Department of Neurology, Esplugues de Llobregat, Barcelona, Spain
| | - Juliana Ribeiro-Constante
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
- Neurometabolic Unit, Hospital Sant Joan de Déu, Department of Neurology, Esplugues de Llobregat, Barcelona, Spain
| | - Xavier Altafaj
- Neurophysiology Laboratory, Department of Biomedicine, Institute of Neurosciences, Faculty of Medicine and Health Sciences, University of Barcelona, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Mireia Olivella
- School of International Studies, ESCI-UPF, Barcelona, Spain
- Bioinformatics and Bioimaging Group, Faculty of Science, Technology and Engineering, University of Vic-Central University of Catalonia, Vic, Spain
| | - Mar O'Callaghan
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
- Neurometabolic Unit, Hospital Sant Joan de Déu, Department of Neurology, Esplugues de Llobregat, Barcelona, Spain
- CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Barcelona, Spain
| | - Alejandra Darling
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
- Neurometabolic Unit, Hospital Sant Joan de Déu, Department of Neurology, Esplugues de Llobregat, Barcelona, Spain
| | - Judith Armstrong
- CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Barcelona, Spain
- Department of Medical Genetics, Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Rafael Artuch
- CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Barcelona, Spain
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Àngels García-Cazorla
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
- Neurometabolic Unit, Hospital Sant Joan de Déu, Department of Neurology, Esplugues de Llobregat, Barcelona, Spain
- CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Barcelona, Spain
| | - Alfonso Oyarzábal
- Synaptic Metabolism and Personalized Therapies Lab, Institut de Recerca Sant Joan de Déu, Department of Neurology and MetabERN, Esplugues de Llobregat, Barcelona, Spain
- Neurometabolic Unit, Hospital Sant Joan de Déu, Department of Neurology, Esplugues de Llobregat, Barcelona, Spain
- CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Barcelona, Spain
| |
Collapse
|
7
|
Smith M, Dodis GE, Vanderplow AM, Gonzalez S, Rhee Y, Gogliotti RG. Potentiation of the M 1 muscarinic acetylcholine receptor normalizes neuronal activation patterns and improves apnea severity in Mecp2+/- mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.586099. [PMID: 38659804 PMCID: PMC11042204 DOI: 10.1101/2024.04.15.586099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is caused by loss-of-function mutations in the methyl-CpG binding protein 2 ( MeCP2 ) gene. RTT patients experience a myriad of debilitating symptoms, which include respiratory phenotypes that are often associated with lethality. Our previous work established that expression of the M 1 muscarinic acetylcholine receptor (mAchR) is decreased in RTT autopsy samples, and that potentiation of the M 1 receptor improves apneas in a mouse model of RTT; however, the population of neurons driving this rescue is unclear. Loss of Mecp2 correlates with excessive neuronal activity in cardiorespiratory nuclei. Since M 1 is found on cholinergic interneurons, we hypothesized that M 1 -potentiating compounds decrease apnea frequency by tempering brainstem hyperactivity. To test this, Mecp2 +/- and Mecp2 +/+ mice were screened for apneas before and after administration of the M 1 positive allosteric modulator (PAM) VU0453595 (VU595). Brains from the same mice were then imaged for c-Fos, ChAT, and Syto16 using whole-brain light-sheet microscopy to establish genotype and drug-dependent activation patterns that could be correlated with VU595's efficacy on apneas. The vehicle-treated Mecp2 +/- brain exhibited broad hyperactivity when coupled with the phenotypic prescreen, which was significantly decreased by administration of VU595, particularly in regions known to modulate the activity of respiratory nuclei (i.e. hippocampus and striatum). Further, the extent of apnea rescue in each mouse showed a significant positive correlation with c-Fos expression in non-cholinergic neurons in the striatum, thalamus, dentate gyrus, and within the cholinergic neurons of the brainstem. These results indicate that Mecp2 +/- mice are prone to hyperactivity in brain regions that regulate respiration, which can be normalized through M 1 potentiation.
Collapse
|
8
|
Parent HH, Niswender CM. Therapeutic Potential for Metabotropic Glutamate Receptor 7 Modulators in Cognitive Disorders. Mol Pharmacol 2024; 105:348-358. [PMID: 38423750 PMCID: PMC11026152 DOI: 10.1124/molpharm.124.000874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is the most highly conserved and abundantly expressed mGlu receptor in the human brain. The presynaptic localization of mGlu7, coupled with its low affinity for its endogenous agonist, glutamate, are features that contribute to the receptor's role in modulating neuronal excitation and inhibition patterns, including long-term potentiation, in various brain regions. These characteristics suggest that mGlu7 modulation may serve as a novel therapeutic strategy in disorders of cognitive dysfunction, including neurodevelopmental disorders that cause impairments in learning, memory, and attention. Primary mutations in the GRM7 gene have recently been identified as novel causes of neurodevelopmental disorders, and these patients exhibit profound intellectual and cognitive disability. Pharmacological tools, such as agonists, antagonists, and allosteric modulators, have been the mainstay for targeting mGlu7 in its endogenous homodimeric form to probe effects of its function and modulation in disease models. However, recent research has identified diversity in dimerization, as well as trans-synaptic interacting proteins, that also play a role in mGlu7 signaling and pharmacological properties. These novel findings represent exciting opportunities in the field of mGlu receptor drug discovery and highlight the importance of further understanding the functions of mGlu7 in complex neurologic conditions at both the molecular and physiologic levels. SIGNIFICANCE STATEMENT: Proper expression and function of mGlu7 is essential for learning, attention, and memory formation at the molecular level within neural circuits. The pharmacological targeting of mGlu7 is undergoing a paradigm shift by incorporating an understanding of receptor interaction with other cis- and trans- acting synaptic proteins, as well as various intracellular signaling pathways. Based upon these new findings, mGlu7's potential as a drug target in the treatment of cognitive disorders and learning impairments is primed for exploration.
Collapse
Affiliation(s)
- Harrison H Parent
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| | - Colleen M Niswender
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| |
Collapse
|
9
|
Mykins M, Bridges B, Jo A, Krishnan K. Multidimensional Analysis of a Social Behavior Identifies Regression and Phenotypic Heterogeneity in a Female Mouse Model for Rett Syndrome. J Neurosci 2024; 44:e1078232023. [PMID: 38199865 PMCID: PMC10957218 DOI: 10.1523/jneurosci.1078-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/01/2023] [Accepted: 11/17/2023] [Indexed: 01/12/2024] Open
Abstract
Regression is a key feature of neurodevelopmental disorders such as autism spectrum disorder, Fragile X syndrome, and Rett syndrome (RTT). RTT is caused by mutations in the X-linked gene methyl-CpG-binding protein 2 (MECP2). It is characterized by an early period of typical development with subsequent regression of previously acquired motor and speech skills in girls. The syndromic phenotypes are individualistic and dynamic over time. Thus far, it has been difficult to capture these dynamics and syndromic heterogeneity in the preclinical Mecp2-heterozygous female mouse model (Het). The emergence of computational neuroethology tools allows for robust analysis of complex and dynamic behaviors to model endophenotypes in preclinical models. Toward this first step, we utilized DeepLabCut, a marker-less pose estimation software to quantify trajectory kinematics and multidimensional analysis to characterize behavioral heterogeneity in Het in the previously benchmarked, ethologically relevant social cognition task of pup retrieval. We report the identification of two distinct phenotypes of adult Het: Het that display a delay in efficiency in early days and then improve over days like wild-type mice and Het that regress and perform worse in later days. Furthermore, regression is dependent on age and behavioral context and can be detected in the initial days of retrieval. Together, the novel identification of two populations of Het suggests differential effects on neural circuitry, opens new avenues to investigate the underlying molecular and cellular mechanisms of heterogeneity, and designs better studies for stratifying therapeutics.
Collapse
Affiliation(s)
- Michael Mykins
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| | - Benjamin Bridges
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| | - Angela Jo
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| | - Keerthi Krishnan
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| |
Collapse
|
10
|
Lei X, Hofmann CS, Rodriguez AL, Niswender CM. Differential Activity of Orthosteric Agonists and Allosteric Modulators at Metabotropic Glutamate Receptor 7. Mol Pharmacol 2023; 104:17-27. [PMID: 37105671 PMCID: PMC10289241 DOI: 10.1124/molpharm.123.000678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is a G protein coupled receptor that has demonstrated promise as a therapeutic target across a number of neurologic and psychiatric diseases. Compounds that modulate the activity of mGlu7, such as positive and negative allosteric modulators, may represent new therapeutic strategies to modulate receptor activity. The endogenous neurotransmitter associated with the mGlu receptor family, glutamate, exhibits low efficacy and potency in activating mGlu7, and surrogate agonists, such as the compound L-(+)-2-Amino-4-phosphonobutyric acid (L-AP4), are often used for receptor activation and compound profiling. To understand the implications of the use of such agonists in the development of positive allosteric modulators (PAMs), we performed a systematic evaluation of receptor activation using a system in which mutations can be made in either protomer of the mGlu7 dimer; we employed mutations that prevent interaction with the orthosteric site as well as the G-protein coupling site of the receptor. We then measured increases in calcium levels downstream of a promiscuous G protein to assess the effects of mutations in one of the two protomers in the presence of two different agonists and three positive allosteric modulators. Our results reveal that distinct PAMs, for example N-[3-Chloro-4-[(5-chloro-2-pyridinyl)oxy]phenyl]-2-pyridinecarboxamide (VU0422288) and 3-(2,3-Difluoro-4-methoxyphenyl)-2,5-dimethyl-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (VU6005649), do exhibit different maximal levels of potentiation with L-AP4 versus glutamate, but there appear to be common stable receptor conformations that are shared among all of the compounds examined here. SIGNIFICANCE STATEMENT: This manuscript describes the systematic evaluation of the mGlu7 agonists glutamate and L-(+)-2-Amino-4-phosphonobutyric acid (L-AP4) in the presence and absence of three distinct potentiators examining possible mechanistic differences. These findings demonstrate that mGlu7 potentiators display subtle variances in response to glutamate versus L-AP4.
Collapse
Affiliation(s)
- Xia Lei
- Department of Pharmacology (X.L., C.S.H., A.L.R., C.M.N.), Warren Center for Neuroscience Drug Discovery (X.L., A.L.R., C.M.N.), Vanderbilt Institute of Chemical Biology (C.M.N.), and Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennesee (C.M.N.); and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| | - Christopher S Hofmann
- Department of Pharmacology (X.L., C.S.H., A.L.R., C.M.N.), Warren Center for Neuroscience Drug Discovery (X.L., A.L.R., C.M.N.), Vanderbilt Institute of Chemical Biology (C.M.N.), and Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennesee (C.M.N.); and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| | - Alice L Rodriguez
- Department of Pharmacology (X.L., C.S.H., A.L.R., C.M.N.), Warren Center for Neuroscience Drug Discovery (X.L., A.L.R., C.M.N.), Vanderbilt Institute of Chemical Biology (C.M.N.), and Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennesee (C.M.N.); and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| | - Colleen M Niswender
- Department of Pharmacology (X.L., C.S.H., A.L.R., C.M.N.), Warren Center for Neuroscience Drug Discovery (X.L., A.L.R., C.M.N.), Vanderbilt Institute of Chemical Biology (C.M.N.), and Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennesee (C.M.N.); and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| |
Collapse
|
11
|
Pershina EV, Chernomorets IY, Fedorov DA, Arkhipov VI. Pharmacological Modulation of Excitotoxicity through the Combined Use of NMDA Receptor Inhibition and Group III mGlu Activation Reduces TMT-Induced Neurodegeneration in the Rat Hippocampus. Int J Mol Sci 2023; 24:ijms24098249. [PMID: 37175959 PMCID: PMC10179112 DOI: 10.3390/ijms24098249] [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: 03/28/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
We studied the neuroprotective properties of the non-competitive NMDA receptor antagonist memantine, in combination with a positive allosteric modulator of metabotropic glutamate receptors of Group III, VU 0422288. The treatment was started 48 h after the injection of neurotoxic agent trimethyltin (TMT) at 7.5 mg/kg. Three weeks after TMT injection, functional and morphological changes in a rat hippocampus were evaluated, including the expression level of genes characterizing glutamate transmission and neuroinflammation, animal behavior, and hippocampal cell morphology. Significant neuronal cell death occurred in the CA3 and CA4 regions, and to a lesser extent, in the CA1 and CA2 regions. The death of neurons in the CA1 field was significantly reduced in animals with a combined use of memantine and VU 0422288. In the hippocampus of these animals, the level of expression of genes characterizing glutamatergic synaptic transmission (Grin2b, Gria1, EAAT2) did not differ from the level in control animals, as well as the expression of genes characterizing neuroinflammation (IL1b, TGF beta 1, Aif1, and GFAP). However, the expression of genes characterizing neuroinflammation was markedly increased in the hippocampus of animals treated with memantine or VU 0422288 alone after TMT. The results of immunohistochemical studies confirmed a significant activation of microglia in the hippocampus three weeks after TMT injection. In contrast to the hilus, microglia in the CA1 region had an increase in rod-like cells. Moreover, in the CA1 field of the hippocampus of the animals of the MEM + VU group, the amount of such microglia was close to the control. Thus, the short-term modulation of glutamatergic synaptic transmission by memantine and subsequent activation of Group III mGluR significantly affected the dynamics of neurodegeneration in the hippocampus.
Collapse
Affiliation(s)
- Ekaterina V Pershina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | - Irina Yu Chernomorets
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | - Dmitry A Fedorov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | - Vladimir I Arkhipov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| |
Collapse
|
12
|
Freitas GA, Niswender CM. GRM7 gene mutations and consequences for neurodevelopment. Pharmacol Biochem Behav 2023; 225:173546. [PMID: 37003303 PMCID: PMC10192299 DOI: 10.1016/j.pbb.2023.173546] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
The metabotropic glutamate receptor 7 (mGlu7), encoded by the GRM7 gene in humans, is a presynaptic, G protein-coupled glutamate receptor that is essential for modulating neurotransmission. Mutations in or reduced expression of GRM7 have been identified in different genetic neurodevelopmental disorders (NDDs), and rare biallelic missense variants have been proposed to underlie a subset of NDDs. Clinical GRM7 variants have been associated with a range of symptoms consistent with neurodevelopmental molecular features, including hypomyelination, brain atrophy and defects in axon outgrowth. Here, we review the newest findings regarding the cellular and molecular defects caused by GRM7 variants in NDD patients.
Collapse
Affiliation(s)
- Geanne A Freitas
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37212, United States of America
| | - Colleen M Niswender
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37212, United States of America; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37212, United States of America; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37212, United States of America; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America.
| |
Collapse
|
13
|
Dickson L, Teall M, Chevalier E, Cheung T, Liwicki GM, Mack S, Stephenson A, White K, Fosbeary R, Harrison DC, Brice NL, Doyle K, Ciccocioppo R, Wu C, Almond S, Patel TR, Mitchell P, Barnes M, Ayscough AP, Dawson LA, Carlton M, Bürli RW. Discovery of CVN636: A Highly Potent, Selective, and CNS Penetrant mGluR 7 Allosteric Agonist. ACS Med Chem Lett 2023; 14:442-449. [PMID: 37077399 PMCID: PMC10107911 DOI: 10.1021/acsmedchemlett.2c00529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
The low affinity metabotropic glutamate receptor mGluR7 has been implicated in numerous CNS disorders; however, a paucity of potent and selective activators has hampered full delineation of the functional role and therapeutic potential of this receptor. In this work, we present the identification, optimization, and characterization of highly potent, novel mGluR7 agonists. Of particular interest is the chromane CVN636, a potent (EC50 7 nM) allosteric agonist which demonstrates exquisite selectivity for mGluR7 compared to not only other mGluRs, but also a broad range of targets. CVN636 demonstrated CNS penetrance and efficacy in an in vivo rodent model of alcohol use disorder. CVN636 thus has potential to progress as a drug candidate in CNS disorders involving mGluR7 and glutamatergic dysfunction.
Collapse
Affiliation(s)
- Louise Dickson
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Martin Teall
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Elodie Chevalier
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Toni Cheung
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Gemma M. Liwicki
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Stephen Mack
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Anne Stephenson
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Kathryn White
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Richard Fosbeary
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - David C. Harrison
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Nicola L. Brice
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Kevin Doyle
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
| | - Chaobo Wu
- WuXi Apptec Limited, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Sarah Almond
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Toshal R. Patel
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Philip Mitchell
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Matt Barnes
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Andrew P. Ayscough
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Lee A. Dawson
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Mark Carlton
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Roland W. Bürli
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| |
Collapse
|
14
|
Design and Synthesis of New Quinazolin-4-one Derivatives with Negative mGlu 7 Receptor Modulation Activity and Antipsychotic-Like Properties. Int J Mol Sci 2023; 24:ijms24031981. [PMID: 36768302 PMCID: PMC9916658 DOI: 10.3390/ijms24031981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 01/21/2023] Open
Abstract
Following the glutamatergic theory of schizophrenia and based on our previous study regarding the antipsychotic-like activity of mGlu7 NAMs, we synthesized a new compound library containing 103 members, which were examined for NAM mGlu7 activity in the T-REx 293 cell line expressing a recombinant human mGlu7 receptor. Out of the twenty-two scaffolds examined, active compounds were found only within the quinazolinone chemotype. 2-(2-Chlorophenyl)-6-(2,3-dimethoxyphenyl)-3-methylquinazolin-4(3H)-one (A9-7, ALX-171, mGlu7 IC50 = 6.14 µM) was selective over other group III mGlu receptors (mGlu4 and mGlu8), exhibited satisfactory drug-like properties in preliminary DMPK profiling, and was further tested in animal models of antipsychotic-like activity, assessing the positive, negative, and cognitive symptoms. ALX-171 reversed DOI-induced head twitches and MK-801-induced disruptions of social interactions or cognition in the novel object recognition test and spatial delayed alternation test. On the other hand, the efficacy of the compound was not observed in the MK-801-induced hyperactivity test or prepulse inhibition. In summary, the observed antipsychotic activity profile of ALX-171 justifies the further development of the group of quinazolin-4-one derivatives in the search for a new drug candidate for schizophrenia treatment.
Collapse
|
15
|
Hoglund BK, Carfagno V, Olive MF, Leyrer-Jackson JM. Metabotropic glutamate receptors and cognition: From underlying plasticity and neuroprotection to cognitive disorders and therapeutic targets. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:367-413. [PMID: 36868635 DOI: 10.1016/bs.irn.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are G protein-coupled receptors that play pivotal roles in mediating the activity of neurons and other cell types within the brain, communication between cell types, synaptic plasticity, and gene expression. As such, these receptors play an important role in a number of cognitive processes. In this chapter, we discuss the role of mGlu receptors in various forms of cognition and their underlying physiology, with an emphasis on cognitive dysfunction. Specifically, we highlight evidence that links mGlu physiology to cognitive dysfunction across brain disorders including Parkinson's disease, Alzheimer's disease, Fragile X syndrome, post-traumatic stress disorder, and schizophrenia. We also provide recent evidence demonstrating that mGlu receptors may elicit neuroprotective effects in particular disease states. Lastly, we discuss how mGlu receptors can be targeted utilizing positive and negative allosteric modulators as well as subtype specific agonists and antagonist to restore cognitive function across these disorders.
Collapse
Affiliation(s)
- Brandon K Hoglund
- Department of Medical Education, School of Medicine, Creighton University, Phoenix, AZ, United States
| | - Vincent Carfagno
- School of Medicine, Midwestern University, Glendale, AZ, United States
| | - M Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Jonna M Leyrer-Jackson
- Department of Medical Education, School of Medicine, Creighton University, Phoenix, AZ, United States.
| |
Collapse
|
16
|
Transgenerational transmission of aspartame-induced anxiety and changes in glutamate-GABA signaling and gene expression in the amygdala. Proc Natl Acad Sci U S A 2022; 119:e2213120119. [PMID: 36459641 PMCID: PMC9894161 DOI: 10.1073/pnas.2213120119] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
We report the effects of aspartame on anxiety-like behavior, neurotransmitter signaling and gene expression in the amygdala, a brain region associated with the regulation of anxiety and fear responses. C57BL/6 mice consumed drinking water containing 0.015% or 0.03% aspartame, a dose equivalent of 8 to 15% of the FDA recommended maximum human daily intake, or plain drinking water. Robust anxiety-like behavior (evaluated using open field test and elevated zero maze) was observed in male and female mice consuming the aspartame-containing water. Diazepam, an allosteric modulator of the GABA-A receptor, alleviated the anxiety-like behavior. RNA sequencing of the amygdala followed by KEGG biological pathway analysis of differentially expressed genes showed glutamatergic and GABAergic synapse pathways as significantly enriched. Quantitative PCR showed upregulation of mRNA for the glutamate NMDA receptor subunit 2D (Grin2d) and metabotropic receptor 4 (Grm4) and downregulation of the GABA-A receptor associated protein (Gabarap) mRNA. Thus, taken together, our diazepam and gene expression data show that aspartame consumption shifted the excitation-inhibition equilibrium in the amygdala toward excitation. Even more strikingly, the anxiety-like behavior, its response to diazepam, and changes in amygdala gene expression were transmitted to male and female offspring in two generations descending from the aspartame-exposed males. Extrapolation of the findings to humans suggests that aspartame consumption at doses below the FDA recommended maximum daily intake may produce neurobehavioral changes in aspartame-consuming individuals and their descendants. Thus, human population at risk of aspartame's potential mental health effects may be larger than current expectations, which only include aspartame-consuming individuals.
Collapse
|
17
|
Frank D, Gruenbaum BF, Zlotnik A, Semyonov M, Frenkel A, Boyko M. Pathophysiology and Current Drug Treatments for Post-Stroke Depression: A Review. Int J Mol Sci 2022; 23:ijms232315114. [PMID: 36499434 PMCID: PMC9738261 DOI: 10.3390/ijms232315114] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Post-stroke depression (PSD) is a biopsychosocial disorder that affects individuals who have suffered a stroke at any point. PSD has a 20 to 60 percent reported prevalence among stroke survivors. Its effects are usually adverse, can lead to disability, and may increase mortality if not managed or treated early. PSD is linked to several other medical conditions, including anxiety, hyper-locomotor activity, and poor functional recovery. Despite significant awareness of its adverse impacts, understanding the pathogenesis of PSD has proved challenging. The exact pathophysiology of PSD is unknown, yet its complexity has been definitively shown, involving mechanisms such as dysfunction of monoamine, the glutamatergic systems, the gut-brain axis, and neuroinflammation. The current effectiveness of PSD treatment is about 30-40 percent of all cases. In this review, we examined different pathophysiological mechanisms and current pharmacological and non-pharmacological approaches for the treatment of PSD.
Collapse
Affiliation(s)
- Dmitry Frank
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion of the Negev, Beer-Sheva 84105, Israel
- Correspondence: or
| | - Benjamin F. Gruenbaum
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Alexander Zlotnik
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion of the Negev, Beer-Sheva 84105, Israel
| | - Michael Semyonov
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion of the Negev, Beer-Sheva 84105, Israel
| | - Amit Frenkel
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion of the Negev, Beer-Sheva 84105, Israel
| | - Matthew Boyko
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
18
|
Lin X, Fisher NM, Dogra S, Senter RK, Reed CW, Kalbfleisch JJ, Lindsley CW, Asher WB, Xiang Z, Niswender CM, Javitch JA. Differential activity of mGlu 7 allosteric modulators provides evidence for mGlu 7/8 heterodimers at hippocampal Schaffer collateral-CA1 synapses. J Biol Chem 2022; 298:102458. [PMID: 36063995 PMCID: PMC9531177 DOI: 10.1016/j.jbc.2022.102458] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
Glutamate acts at eight metabotropic glutamate (mGlu) receptor subtypes expressed in a partially overlapping fashion in distinct brain circuits. Recent evidence indicates that specific mGlu receptor protomers can heterodimerize and that these heterodimers can exhibit different pharmacology when compared to their homodimeric counterparts. Group III mGlu agonist-induced suppression of evoked excitatory potentials and induction of long-term potentiation at Schaffer collateral-CA1 (SC-CA1) synapses in the rodent hippocampus can be blocked by the selective mGlu7 negative allosteric modulator (NAM), ADX71743. Curiously, a different mGlu7 NAM, 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazonolo[4,5-c]pyridin-4(5H)-one, failed to block these responses in brain slices despite its robust activity at mGlu7 homodimers in vitro. We hypothesized that this might result from heterodimerization of mGlu7 with another mGlu receptor protomer and focused on mGlu8 as a candidate given the reported effects of mGlu8-targeted compounds in the hippocampus. Here, we used complemented donor acceptor-resonance energy transfer to study mGlu7/8 heterodimer activation in vitro and observed that ADX71743 blocked responses of both mGlu7/7 homodimers and mGlu7/8 heterodimers, whereas 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazonolo[4,5-c]pyridin-4(5H)-one only antagonized responses of mGlu7/7 homodimers. Taken together with our electrophysiology observations, these results suggest that a receptor with pharmacology consistent with an mGlu7/8 heterodimer modulates the activity of SC-CA1 synapses. Building on this hypothesis, we identified two additional structurally related mGlu7 NAMs that also differ in their activity at mGlu7/8 heterodimers, in a manner consistent with their ability to inhibit synaptic transmission and plasticity at SC-CA1. Thus, we propose that mGlu7/8 heterodimers are a key molecular target for modulating the activity of hippocampal SC-CA1 synapses.
Collapse
Affiliation(s)
- Xin Lin
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Shalini Dogra
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Rebecca K Senter
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Carson W Reed
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Jacob J Kalbfleisch
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA; Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Wesley B Asher
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Zixiu Xiang
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA; Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| | - Jonathan A Javitch
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA.
| |
Collapse
|
19
|
Witkin JM, Pandey KP, Smith JL. Clinical investigations of compounds targeting metabotropic glutamate receptors. Pharmacol Biochem Behav 2022; 219:173446. [PMID: 35987339 DOI: 10.1016/j.pbb.2022.173446] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/22/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Pharmacological modulation of glutamate has long been considered to be of immense therapeutic utility. The metabotropic glutamate receptors (mGluRs) are potential targets for safely altering glutamate-driven excitation. Data support the potential therapeutic use of mGluR modulators in the treatment of anxiety, depression, schizophrenia, and other psychiatric disorders, pain, epilepsy, as well as neurodegenerative and neurodevelopmental disorders. For each of the three mGluR groups, compounds have been constructed that produce either potentiation or functional blockade. PET ligands for mGlu5Rs have been studied in a range of patient populations and several mGlu5R antagonists have been tested for potential efficacy in patients including mavoglurant, diploglurant, basimglurant, GET 73, and ADX10059. Efficacy with mGlu5R antagonists has been reported in trials with patients with gastroesophageal reflux disease; data from patients with Parkinson's disease or Fragile X syndrome have not been as robust as hoped. Fenobam was approved for use as an anxiolytic prior to its recognition as an mGlu5R antagonist. mGlu2/3R agonists (pomaglumated methionil) and mGlu2R agonists (JNJ-40411813, AZD 8529, and LY2979165) have been studied in patients with schizophrenia with promising but mixed results. Antagonists of mGlu2/3Rs (decoglurant and TS-161) have been studied in depression where TS-161 has advanced into a planned Phase 2 study in treatment-resistant depression. The Group III mGluRs are the least developed of the mGluR receptor targets. The mGlu4R potentiator, foliglurax, did not meet its primary endpoint in patients with Parkinson's disease. Ongoing efforts to develop mGluR-targeted compounds continue to promise these glutamate modulators as medicines for psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA; Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | - Kamal P Pandey
- Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA
| |
Collapse
|
20
|
The mGlu 7 receptor in schizophrenia - An update and future perspectives. Pharmacol Biochem Behav 2022; 218:173430. [PMID: 35870668 DOI: 10.1016/j.pbb.2022.173430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022]
Abstract
The mGlu7 receptor belongs to the III group of metabotropic glutamatergic (mGlu) receptors and physiologically serves as an "emergency" receptor that is activated by high, almost pathological, glutamate concentrations. Of all mGlu receptors, this receptor is most highly expressed in the brain. Additionally, relatively intense expression of the receptor was found at the periphery, for example in the bowels or in the reproductive system of male mice, but this review will be focused predominantly on its role in the brain. In the CNS, the receptor is expressed presynaptically, in the center of the synaptic cleft, at the terminals of both excitatory glutamatergic and inhibitory GABAergic neurons. Thus, it may regulate the release of both glutamate and GABA. Schizophrenia is thought to develop as a consequence of a disturbed glutamatergic-GABAergic balance in different parts of the brain. Thus, the mGlu7 receptor may be involved in the pathophysiology of schizophrenia and consequently constitute the target for antipsychotic drug discovery. In this review, we summarize the available data about mGlu7 receptor ligands and their activity in animal models of schizophrenia. At present, only a few ligands are available, and negative allosteric modulators (NAMs) appear to exert antipsychotic-like efficacy, indicating that the inhibition of the receptor could constitute a promising target in the search for novel drugs. Additionally, the data concerning the expression of the receptor in the CNS and putative mechanisms by which its inhibition may contribute to the treatment of schizophrenia will be discussed. Finally, the polymorphisms of genes encoding the receptor in schizophrenic patients will also be provided.
Collapse
|
21
|
Xu P, Yue Y, Su J, Sun X, Du H, Liu Z, Simha R, Zhou J, Zeng C, Lu H. Pattern decorrelation in the mouse medial prefrontal cortex enables social preference and requires MeCP2. Nat Commun 2022; 13:3899. [PMID: 35794118 PMCID: PMC9259602 DOI: 10.1038/s41467-022-31578-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 06/21/2022] [Indexed: 11/08/2022] Open
Abstract
Sociability is crucial for survival, whereas social avoidance is a feature of disorders such as Rett syndrome, which is caused by loss-of-function mutations in MECP2. To understand how a preference for social interactions is encoded, we used in vivo calcium imaging to compare medial prefrontal cortex (mPFC) activity in female wild-type and Mecp2-heterozygous mice during three-chamber tests. We found that mPFC pyramidal neurons in Mecp2-deficient mice are hypo-responsive to both social and nonsocial stimuli. Hypothesizing that this limited dynamic range restricts the circuit's ability to disambiguate coactivity patterns for different stimuli, we suppressed the mPFC in wild-type mice and found that this eliminated both pattern decorrelation and social preference. Conversely, stimulating the mPFC in MeCP2-deficient mice restored social preference, but only if it was sufficient to restore pattern decorrelation. A loss of social preference could thus indicate impaired pattern decorrelation rather than true social avoidance.
Collapse
Affiliation(s)
- Pan Xu
- The GW Institute for Neuroscience, The George Washington University, Washington, DC, 20037, USA
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20037, USA
- Institute of Basic Science, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China
| | - Yuanlei Yue
- The GW Institute for Neuroscience, The George Washington University, Washington, DC, 20037, USA
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20037, USA
| | - Juntao Su
- The GW Institute for Neuroscience, The George Washington University, Washington, DC, 20037, USA
| | - Xiaoqian Sun
- Department of Computer Science, School of Engineering and Applied Science, The George Washington University, Washington, DC, 20037, USA
| | - Hongfei Du
- Department of Statistics, Columbian College of Art and Sciences, The George Washington University, Washington, DC, 20037, USA
| | - Zhichao Liu
- Department of Physics, Columbian College of Art and Sciences, The George Washington University, Washington, DC, 20037, USA
- School of Biological Information, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China
| | - Rahul Simha
- Department of Computer Science, School of Engineering and Applied Science, The George Washington University, Washington, DC, 20037, USA
| | - Jianhui Zhou
- Department of Statistics, School of Arts and Sciences, University of Virginia, Charlottesville, VA, 22904, USA
| | - Chen Zeng
- Department of Statistics, Columbian College of Art and Sciences, The George Washington University, Washington, DC, 20037, USA
| | - Hui Lu
- The GW Institute for Neuroscience, The George Washington University, Washington, DC, 20037, USA.
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20037, USA.
| |
Collapse
|
22
|
Smith M, Arthur B, Cikowski J, Holt C, Gonzalez S, Fisher NM, Vermudez SAD, Lindsley CW, Niswender CM, Gogliotti RG. Clinical and Preclinical Evidence for M 1 Muscarinic Acetylcholine Receptor Potentiation as a Therapeutic Approach for Rett Syndrome. Neurotherapeutics 2022; 19:1340-1352. [PMID: 35670902 PMCID: PMC9587166 DOI: 10.1007/s13311-022-01254-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is characterized by developmental regression, loss of communicative ability, stereotyped hand wringing, cognitive impairment, and central apneas, among many other symptoms. RTT is caused by loss-of-function mutations in a methyl-reader known as methyl-CpG-binding protein 2 (MeCP2), a protein that links epigenetic changes on DNA to larger chromatin structure. Historically, target identification for RTT has relied heavily on Mecp2 knockout mice; however, we recently adopted the alternative approach of performing transcriptional profiling in autopsy samples from RTT patients. Through this mechanism, we identified muscarinic acetylcholine receptors (mAChRs) as potential therapeutic targets. Here, we characterized a cohort of 40 temporal cortex samples from individuals with RTT and quantified significantly decreased levels of the M1, M2, M3, and M5 mAChRs subtypes relative to neurotypical controls. Of these four subtypes, M1 expression demonstrated a linear relationship with MeCP2 expression, such that M1 levels were only diminished in contexts where MeCP2 was also significantly decreased. Further, we show that M1 potentiation with the positive allosteric modulator (PAM) VU0453595 (VU595) rescued social preference, spatial memory, and associative memory deficits, as well as decreased apneas in Mecp2+/- mice. VU595's efficacy on apneas in Mecp2+/- mice was mediated by the facilitation of the transition from inspiration to expiration. Molecular analysis correlated rescue with normalized global gene expression patterns in the brainstem and hippocampus, as well as increased Gsk3β inhibition and NMDA receptor trafficking. Together, these data suggest that M1 PAMs could represent a new class of RTT therapeutics.
Collapse
Affiliation(s)
- Mackenzie Smith
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Bright Arthur
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jakub Cikowski
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Calista Holt
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Sonia Gonzalez
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sheryl Anne D Vermudez
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - Rocco G Gogliotti
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA.
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
23
|
Cikowski J, Holt C, Arthur B, Smith M, Gonzalez S, Lindsley CW, Niswender CM, Gogliotti RG. Optimized Administration of the M 4 PAM VU0467154 Demonstrates Broad Efficacy, but Limited Effective Concentrations in Mecp2+/- Mice. ACS Chem Neurosci 2022; 13:1891-1901. [PMID: 35671352 PMCID: PMC9266622 DOI: 10.1021/acschemneuro.2c00113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Hypofunction of cholinergic circuits and diminished cholinergic tone have been associated with the neurodevelopmental disorder Rett syndrome (RTT). Specifically, deletion of Mecp2 in cholinergic neurons evokes the same social and cognitive phenotypes in mice seen with global Mecp2 knockout, and decreased choline acetyltransferase activity and vesamicol binding have been reported in RTT autopsy samples. Further, we recently identified significant decreases in muscarinic acetylcholine receptor subtype 4 (M4) expression in both the motor cortex and cerebellum of RTT patient autopsies and established proof of concept that an acute dose of the positive allosteric modulator (PAM) VU0467154 (VU154) rescued phenotypes in Mecp2+/- mice. Here, we expand the assessment of M4 PAMs in RTT to address clinically relevant questions of tolerance, scope of benefit, dose response, chronic treatment, and mechanism. We show that VU154 has efficacy on anxiety, social preference, cognitive, and respiratory phenotypes in Mecp2+/- mice; however, the therapeutic range is narrow, with benefits seen at 3 mg/kg concentrations, but not 1 or 10 mg/kg. Further, sociability was diminished in VU154-treated Mecp2+/- mice, suggestive of a potential adverse effect. Compound efficacy on social, cognitive, and respiratory phenotypes was conserved with a 44-day treatment paradigm, with the caveat that breath rate was moderately decreased with chronic treatment in Mecp2+/+ and Mecp2+/- mice. VU154 effects on respiratory function correlated with an increase in Gsk3β inhibition in the brainstem. These results identify the core symptom domains where efficacy and adverse effects may present with M4 administration in RTT model mice and advocate for the continued evaluation as potential RTT therapeutics.
Collapse
Affiliation(s)
- Jakub Cikowski
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States
| | - Calista Holt
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States
| | - Bright Arthur
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States,Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Mackenzie Smith
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States
| | - Sonia Gonzalez
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States
| | - Craig W. Lindsley
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States,Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States,Vanderbilt
Institute of Chemical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States,Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Colleen M. Niswender
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States,Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States,Vanderbilt
Institute of Chemical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States,Vanderbilt
Brain Institute, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Rocco G. Gogliotti
- Department
of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois 60153, United States,Edward
Hines Jr. VA Hospital, Hines, Illinois 60141, United States,Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States,Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,. Phone: 708-216-9021. Fax: 708-216-8318
| |
Collapse
|
24
|
Exploration of group II metabotropic glutamate receptor modulation in mouse models of Rett syndrome and MECP2 Duplication syndrome. Neuropharmacology 2022; 209:109022. [PMID: 35248529 PMCID: PMC8973998 DOI: 10.1016/j.neuropharm.2022.109022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/11/2022] [Accepted: 02/27/2022] [Indexed: 01/01/2023]
Abstract
Rett syndrome (RTT) and MECP2 Duplication syndrome (MDS) have opposing molecular origins in relation to expression and function of the transcriptional regulator Methyl-CpG-binding protein 2 (MeCP2). Several clinical and preclinical phenotypes, however, are shared between these disorders. Modulation of MeCP2 levels has recently emerged as a potential treatment option for both of these diseases. However, toxicity concerns remain with these approaches. Here, we focus on pharmacologically modulating the group II metabotropic glutamate receptors (mGlu), mGlu2 and mGlu3, which are two downstream targets of MeCP2 that are bidirectionally affected in expression in RTT patients and mice (Mecp2Null/+) versus an MDS mouse model (MECP2Tg1/o). Mecp2Null/+ and MECP2Tg1/o animals also exhibit contrasting phenotypes in trace fear acquisition, a form of temporal associative learning and memory, with trace fear deficiency observed in Mecp2Null/+ mice and abnormally enhanced trace fear acquisition in MECP2Tg1/o animals. In Mecp2Null/+ mice, treatment with the mGlu2/3 agonist LY379268 reverses the deficit in trace fear acquisition, and mGlu2/3 antagonism with LY341495 normalizes the abnormal trace fear learning and memory phenotype in MECP2Tg1/o mice. Altogether, these data highlight the role of group II mGlu receptors in RTT and MDS and demonstrate that both mGlu2 and mGlu3 may be potential therapeutic targets for these disorders.
Collapse
|
25
|
Li W. Excitation and Inhibition Imbalance in Rett Syndrome. Front Neurosci 2022; 16:825063. [PMID: 35250460 PMCID: PMC8894599 DOI: 10.3389/fnins.2022.825063] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
A loss of the excitation/inhibition (E/I) balance in the neural circuit has emerged as a common neuropathological feature in many neurodevelopmental disorders. Rett syndrome (RTT), a prevalent neurodevelopmental disorder that affects 1:10,000-15,000 women globally, is caused by loss-of-function mutations in the Methyl-CpG-binding Protein-2 (Mecp2) gene. E/I imbalance is recognized as the leading cellular and synaptic hallmark that is fundamental to diverse RTT neurological symptoms, including stereotypic hand movements, impaired motor coordination, breathing irregularities, seizures, and learning/memory dysfunctions. E/I balance in RTT is not homogeneously altered but demonstrates brain region and cell type specificity instead. In this review, I elaborate on the current understanding of the loss of E/I balance in a range of brain areas at molecular and cellular levels. I further describe how the underlying cellular mechanisms contribute to the disturbance of the proper E/I ratio. Last, I discuss current pharmacologic innervations for RTT and their role in modifying the E/I balance.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
26
|
Collins BE, Merritt JK, Erickson KR, Neul JL. Safety and efficacy of genetic MECP2 supplementation in the R294X mouse model of Rett syndrome. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12739. [PMID: 33942492 PMCID: PMC8563491 DOI: 10.1111/gbb.12739] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 01/03/2023]
Abstract
Rett syndrome is a neurodevelopmental disorder caused predominantly by loss-of-function mutations in MECP2, encoding transcriptional modulator methyl-CpG-binding protein 2 (MeCP2). Although no disease-modifying therapies exist at this time, some proposed therapeutic strategies aim to supplement the mutant allele with a wild-type allele producing typical levels of functional MeCP2, such as gene therapy. Because MECP2 is a dosage-sensitive gene, with both loss and gain of function causing disease, these approaches must achieve a narrow therapeutic window to be both safe and effective. While MeCP2 supplementation rescues RTT-like phenotypes in mouse models, the tolerable threshold of MeCP2 is not clear, particularly for partial loss-of-function mutations. We assessed the safety of genetically supplementing full-length human MeCP2 in the context of the R294X allele, a common partial loss-of-function mutation retaining DNA-binding capacity. We assessed the potential for adverse effects from MeCP2 supplementation of a partial loss-of-function mutant and the potential for dominant negative interactions between mutant and full-length MeCP2. In male hemizygous R294X mice, MeCP2 supplementation rescued RTT-like behavioral phenotypes and did not elicit behavioral evidence of excess MeCP2. In female heterozygous R294X mice, RTT-specific phenotypes were similarly rescued. However, MeCP2 supplementation led to evidence of excess MeCP2 activity in a motor coordination assay, suggesting that the underlying motor circuitry is particularly sensitive to MeCP2 dosage in females. These results show that genetic supplementation of full-length MeCP2 is safe in males and largely so females. However, careful consideration of risk for adverse motor effects may be warranted for girls and women with RTT.
Collapse
Affiliation(s)
| | - Jonathan K. Merritt
- Department of PediatricsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Kirsty R. Erickson
- Department of PediatricsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jeffrey L. Neul
- Vanderbilt Kennedy Center, Departments of Pediatrics, Pharmacology, and Special EducationVanderbilt University Medical Center and Vanderbilt UniversityNashvilleTennesseeUSA
| |
Collapse
|
27
|
Singh J, Lanzarini E, Nardocci N, Santosh P. Movement disorders in patients with Rett syndrome: A systematic review of evidence and associated clinical considerations. Psychiatry Clin Neurosci 2021; 75:369-393. [PMID: 34472659 PMCID: PMC9298304 DOI: 10.1111/pcn.13299] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 08/20/2021] [Indexed: 12/18/2022]
Abstract
AIM This systematic review identified and thematically appraised clinical evidence of movement disorders in patients with Rett syndrome (RTT). METHOD Using PRISMA criteria, six electronic databases were searched from inception to April 2021. A thematic analysis was then undertaken on the extracted data to identify potential themes. RESULTS Following the thematic analysis, six themes emerged: (i) clinical features of abnormal movement behaviors; (ii) mutational profile and its impact on movement disorders; (iii) symptoms and stressors that impact on movement disorders; (iv) possible underlying neurobiological mechanisms; (v) quality of life and movement disorders; and (vi) treatment of movement disorders. Current guidelines for managing movement disorders in general were then reviewed to provide possible treatment recommendations for RTT. CONCLUSION Our study offers an enriched data set for clinical investigations and treatment of fine and gross motor issues in RTT. A detailed understanding of genotype-phenotype relationships of movement disorders allows for more robust genetic counseling for families but can also assist healthcare professionals in terms of monitoring disease progression in RTT. The synthesis also showed that environmental enrichment would be beneficial for improving some aspects of movement disorders. The cerebellum, basal ganglia, alongside dysregulation of the cortico-basal ganglia-thalamo-cortical loop, are likely anatomical targets. A review of treatments for movement disorders also helped to provide recommendations for treating and managing movement disorders in patients with RTT.
Collapse
Affiliation(s)
- Jatinder Singh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Interventional Paediatric Psychopharmacology and Rare Diseases, South London and Maudsley NHS Foundation Trust, London, UK.,Centre for Personalised Medicine in Rett Syndrome, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Evamaria Lanzarini
- Child and Adolescent Neuropsychiatry Unit, Infermi Hospital, Rimini, Italy
| | - Nardo Nardocci
- Department of Paediatric Neurology, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Paramala Santosh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Interventional Paediatric Psychopharmacology and Rare Diseases, South London and Maudsley NHS Foundation Trust, London, UK.,Centre for Personalised Medicine in Rett Syndrome, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
28
|
Vermudez SAD, Gogliotti RG, Arthur B, Buch A, Morales C, Moxley Y, Rajpal H, Conn PJ, Niswender CM. Profiling beneficial and potential adverse effects of MeCP2 overexpression in a hypomorphic Rett syndrome mouse model. GENES, BRAIN, AND BEHAVIOR 2021; 21:e12752. [PMID: 34002468 PMCID: PMC8599502 DOI: 10.1111/gbb.12752] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 01/03/2023]
Abstract
De novo loss-of-function mutations in methyl-CpG-binding protein 2 (MeCP2) lead to the neurodevelopmental disorder Rett syndrome (RTT). Despite promising results from strategies aimed at increasing MeCP2 levels, additional studies exploring how hypomorphic MeCP2 mutations impact the therapeutic window are needed. Here, we investigated the consequences of genetically introducing a wild-type MECP2 transgene in the Mecp2 R133C mouse model of RTT. The MECP2 transgene reversed the majority of RTT-like phenotypes exhibited by male and female Mecp2 R133C mice. However, three core symptom domains were adversely affected in female Mecp2R133C/+ animals; these phenotypes resemble those observed in disease contexts of excess MeCP2. Parallel control experiments in Mecp2Null/+ mice linked these adverse effects to the hypomorphic R133C mutation. Collectively, these data provide evidence regarding the safety and efficacy of genetically overexpressing functional MeCP2 in Mecp2 R133C mice and suggest that personalized approaches may warrant consideration for the clinical assessment of MeCP2-targeted therapies.
Collapse
Affiliation(s)
- Sheryl Anne D. Vermudez
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Rocco G. Gogliotti
- Department of Molecular Pharmacology and NeuroscienceLoyola University ChicagoChicagoIllinoisUSA
| | - Bright Arthur
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Aditi Buch
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Clarissa Morales
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Yuta Moxley
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Hemangi Rajpal
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - P. Jeffrey Conn
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA,Vanderbilt Kennedy CenterVanderbilt UniversityNashvilleTennesseeUSA,Vanderbilt Institute of Chemical BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Colleen M. Niswender
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA,Vanderbilt Kennedy CenterVanderbilt UniversityNashvilleTennesseeUSA,Vanderbilt Institute of Chemical BiologyVanderbilt UniversityNashvilleTennesseeUSA
| |
Collapse
|
29
|
Fisher NM, AlHashim A, Buch AB, Badivuku H, Samman MM, Weiss KM, Cestero GI, Does MD, Rook JM, Lindsley CW, Conn PJ, Gogliotti RG, Niswender CM. A GRM7 mutation associated with developmental delay reduces mGlu7 expression and produces neurological phenotypes. JCI Insight 2021; 6:143324. [PMID: 33476302 PMCID: PMC7934925 DOI: 10.1172/jci.insight.143324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/13/2021] [Indexed: 12/29/2022] Open
Abstract
The metabotropic glutamate receptor 7 (mGlu7) is a G protein–coupled receptor that has been recently linked to neurodevelopmental disorders. This association is supported by the identification of GRM7 variants in patients with autism spectrum disorder, attention deficit hyperactivity disorder, and severe developmental delay. One GRM7 mutation previously reported in 2 patients results in a single amino acid change, I154T, within the mGlu7 ligand-binding domain. Here, we report 2 new patients with this mutation who present with severe developmental delay and epilepsy. Functional studies of the mGlu7-I154T mutant reveal that this substitution resulted in significant loss of mGlu7 protein expression in HEK293A cells and in mice. We show that this occurred posttranscriptionally at the level of protein expression and trafficking. Similar to mGlu7–global KO mice, mGlu7-I154T animals exhibited reduced motor coordination, deficits in contextual fear learning, and seizures. This provides functional evidence that a disease-associated mutation affecting the mGlu7 receptor was sufficient to cause neurological dysfunction in mice and further validates GRM7 as a disease-causing gene in the human population.
Collapse
Affiliation(s)
- Nicole M Fisher
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Aditi B Buch
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Hana Badivuku
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Kelly M Weiss
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Gabriela I Cestero
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Mark D Does
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jerri M Rook
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Craig W Lindsley
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Department of Chemistry and.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - P Jeffrey Conn
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee USA
| | - Rocco G Gogliotti
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois, USA
| | - Colleen M Niswender
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee USA
| |
Collapse
|
30
|
Pathogenic GRM7 Mutations Associated with Neurodevelopmental Disorders Impair Axon Outgrowth and Presynaptic Terminal Development. J Neurosci 2021; 41:2344-2359. [PMID: 33500274 DOI: 10.1523/jneurosci.2108-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 01/11/2021] [Accepted: 01/16/2021] [Indexed: 12/18/2022] Open
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is an inhibitory heterotrimeric G-protein-coupled receptor that modulates neurotransmitter release and synaptic plasticity at presynaptic terminals in the mammalian central nervous system. Recent studies have shown that rare mutations in glutamate receptors and synaptic scaffold proteins are associated with neurodevelopmental disorders (NDDs). However, the role of presynaptic mGlu7 in the pathogenesis of NDDs remains largely unknown. Recent whole-exome sequencing (WES) studies in families with NDDs have revealed that several missense mutations (c.1865G>A:p.R622Q; c.461T>C:p.I154T; c.1972C>T:p.R658W and c.2024C>A:p.T675K) or a nonsense mutation (c.1757G>A:p.W586X) in the GRM7 gene may be linked to NDDs. In the present study, we investigated the mechanistic links between GRM7 point mutations and NDD pathology. We find that the pathogenic GRM7 I154T and R658W/T675K mutations lead to the degradation of the mGlu7 protein. In particular, the GRM7 R658W/T675K mutation results in a lack of surface mGlu7 expression in heterologous cells and cultured neurons isolated from male and female rat embryos. We demonstrate that the expression of mGlu7 variants or exposure to mGlu7 antagonists impairs axon outgrowth through the mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling pathway during early neuronal development, which subsequently leads to a decrease in the number of presynaptic terminals in mature neurons. Treatment with an mGlu7 agonist restores the pathologic phenotypes caused by mGlu7 I154T but not by mGlu7 R658W/T675K because of its lack of neuronal surface expression. These findings provide evidence that stable neuronal surface expression of mGlu7 is essential for neural development and that mGlu7 is a promising therapeutic target for NDDs.SIGNIFICANCE STATEMENT Neurodevelopmental disorders (NDDs) affect brain development and function by multiple etiologies. Metabotropic glutamate receptor 7 (mGlu7) is a receptor that controls excitatory neurotransmission and synaptic plasticity. Since accumulating evidence indicates that the GRM7 gene locus is associated with NDD risk, we analyzed the functional effects of human GRM7 variants identified in patients with NDDs. We demonstrate that stable neuronal surface expression of mGlu7 is essential for axon outgrowth and presynaptic terminal development in neurons. We found that mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling and subsequent cytoskeletal dynamics are defective because of the degradation of mGlu7 variants. Finally, we show that the defects caused by mGlu7 I154T can be reversed by agonists, providing the rationale for proposing mGlu7 as a potential therapeutic target for NDDs.
Collapse
|
31
|
Gregory KJ, Goudet C. International Union of Basic and Clinical Pharmacology. CXI. Pharmacology, Signaling, and Physiology of Metabotropic Glutamate Receptors. Pharmacol Rev 2020; 73:521-569. [PMID: 33361406 DOI: 10.1124/pr.119.019133] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors respond to glutamate, the major excitatory neurotransmitter in the mammalian brain, mediating a modulatory role that is critical for higher-order brain functions such as learning and memory. Since the first mGlu receptor was cloned in 1992, eight subtypes have been identified along with many isoforms and splice variants. The mGlu receptors are transmembrane-spanning proteins belonging to the class C G protein-coupled receptor family and represent attractive targets for a multitude of central nervous system disorders. Concerted drug discovery efforts over the past three decades have yielded a wealth of pharmacological tools including subtype-selective agents that competitively block or mimic the actions of glutamate or act allosterically via distinct sites to enhance or inhibit receptor activity. Herein, we review the physiologic and pathophysiological roles for individual mGlu receptor subtypes including the pleiotropic nature of intracellular signal transduction arising from each. We provide a comprehensive analysis of the in vitro and in vivo pharmacological properties of prototypical and commercially available orthosteric agonists and antagonists as well as allosteric modulators, including ligands that have entered clinical trials. Finally, we highlight emerging areas of research that hold promise to facilitate rational design of highly selective mGlu receptor-targeting therapeutics in the future. SIGNIFICANCE STATEMENT: The metabotropic glutamate receptors are attractive therapeutic targets for a range of psychiatric and neurological disorders. Over the past three decades, intense discovery efforts have yielded diverse pharmacological tools acting either competitively or allosterically, which have enabled dissection of fundamental biological process modulated by metabotropic glutamate receptors and established proof of concept for many therapeutic indications. We review metabotropic glutamate receptor molecular pharmacology and highlight emerging areas that are offering new avenues to selectively modulate neurotransmission.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| | - Cyril Goudet
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| |
Collapse
|
32
|
Li W, Kutas M, Gray JA, Hagerman RH, Olichney JM. The Role of Glutamate in Language and Language Disorders - Evidence from ERP and Pharmacologic Studies. Neurosci Biobehav Rev 2020; 119:217-241. [PMID: 33039453 PMCID: PMC11584167 DOI: 10.1016/j.neubiorev.2020.09.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 08/10/2020] [Accepted: 09/21/2020] [Indexed: 12/31/2022]
Abstract
Current models of language processing do not address mechanisms at the neurotransmitter level, nor how pharmacologic agents may improve language function(s) in seemingly disparate disorders. L-Glutamate, the primary excitatory neurotransmitter in the human brain, is extensively involved in various higher cortical functions. We postulate that the physiologic role of L-Glutamate neurotransmission extends to the regulation of language access, comprehension, and production, and that disorders in glutamatergic transmission and circuitry contribute to the pathogenesis of neurodegenerative diseases and sporadic-onset language disorders such as the aphasic stroke syndromes. We start with a review of basic science data pertaining to various glutamate receptors in the CNS and ways that they may influence the physiological processes of language access and comprehension. We then focus on the dysregulation of glutamate neurotransmission in three conditions in which language dysfunction is prominent: Alzheimer's Disease, Fragile X-associated Tremor/Ataxia Syndrome, and Aphasic Stroke Syndromes. Finally, we review the pharmacologic and electrophysiologic (event related brain potential or ERP) data pertaining to the role glutamate neurotransmission plays in language processing and disorders.
Collapse
Affiliation(s)
- Wentao Li
- Department of Neurology, University of California, Davis, 4860 Y Street, Suite 3700, Sacramento, CA, 95817, USA.
| | - Marta Kutas
- Department of Cognitive Science, University of California, San Diego, 9500 Gilman Drive #0515, La Jolla, CA, 92093, USA; Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - John A Gray
- Department of Neurology, University of California, Davis, 4860 Y Street, Suite 3700, Sacramento, CA, 95817, USA; Center for Neuroscience, University of California, Davis, 1544 Newton Court, Davis, CA, 95618, USA.
| | - Randi H Hagerman
- MIND Institute, University of California, Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| | - John M Olichney
- Department of Neurology, University of California, Davis, 4860 Y Street, Suite 3700, Sacramento, CA, 95817, USA; Center for Mind and Brain, University of California, Davis, 267 Cousteau Place, Davis, CA, 95618, USA.
| |
Collapse
|
33
|
Kalbfleisch JJ, Reed CW, Park C, Spearing PK, Quitalig MC, Jenkins MT, Rodriguez AL, Blobaum AL, Conn PJ, Niswender CM, Lindsley CW. Synthesis and SAR of a series of mGlu 7 NAMs based on an ethyl-8-methoxy-4-(4-phenylpiperazin-1-yl)quinoline carboxylate core. Bioorg Med Chem Lett 2020; 30:127529. [PMID: 32890686 PMCID: PMC7686273 DOI: 10.1016/j.bmcl.2020.127529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 10/23/2022]
Abstract
A High-Throughput Screening (HTS) campaign identified a fundamentally new mGlu7 NAM chemotype, based on an ethyl-8-methoxy-4-(4-phenylpiperazin-1-yl)quinolone carboxylate core. The initial hit, VU0226390, was a potent mGlu7 NAM (IC50 = 647 nM, 6% L-AP4 min) with selectivity versus the other group III mGlu receptors (>30 μM vs. mGlu4 and mGlu8). A multi-dimensional optimization effort surveyed all regions of this new chemotype, and found very steep SAR, reminiscent of allosteric modulators, and unexpected piperazine mimetics (whereas classical bioisosteres failed). While mGlu7 NAM potency could be improved (IC50s ~ 350 nM), the necessity of the ethyl ester moiety and poor physiochemical and DMPK properties precluded optimization towards in vivo tool compounds or clinical candidates. Still, this hit-to-lead campaign afforded key medicinal chemistry insights and new opportunities.
Collapse
Affiliation(s)
- Jacob J Kalbfleisch
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Carson W Reed
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Charlotte Park
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Paul K Spearing
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Marc C Quitalig
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Matthew T Jenkins
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alice L Rodriguez
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Anna L Blobaum
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA.
| | - Craig W Lindsley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
34
|
Gomathi M, Padmapriya S, Balachandar V. Drug Studies on Rett Syndrome: From Bench to Bedside. J Autism Dev Disord 2020; 50:2740-2764. [PMID: 32016693 DOI: 10.1007/s10803-020-04381-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Drug studies on Rett syndrome (RTT) have drastically increased over the past few decades. This review aims to provide master data on bench-to-bedside drug studies involving RTT. A comprehensive literature review was performed by searching in PUBMED, MEDLINE and Google Scholar, international, national and regional clinical trial registries and pharmaceutical companies using the keywords "Rett syndrome treatment and/or drug or compound or molecule". Seventy drugs were investigated in non-clinical (N = 65 animal/cell line-based studies; N = 5 iPSC-based study) and clinical trials (N = 34) for ameliorating the symptoms of RTT. Though there is good progress in both clinical and non-clinical studies, none of these drugs entered phase III/IV for being launched as a therapeutic agent for RTT.
Collapse
Affiliation(s)
- Mohan Gomathi
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | | | - Vellingiri Balachandar
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India.
| |
Collapse
|
35
|
Reed C, Kalbfleisch JJ, Wong MJ, Washecheck JP, Hunter A, Rodriguez AL, Blobaum AL, Conn PJ, Niswender CM, Lindsley CW. Discovery of VU6027459: A First-in-Class Selective and CNS Penetrant mGlu 7 Positive Allosteric Modulator Tool Compound. ACS Med Chem Lett 2020; 11:1773-1779. [PMID: 32944146 PMCID: PMC7488291 DOI: 10.1021/acsmedchemlett.0c00432] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/20/2020] [Indexed: 01/07/2023] Open
Abstract
Herein, we report the discovery of the first selective and CNS penetrant mGlu7 PAM (VU6027459) derived from a "molecular switch" within a selective mGlu7 NAM chemotype. VU6027459 displayed CNS penetration in both mice (Kp = 2.74) and rats (Kp= 4.78), it was orally bioavailable in rats (%F = 69.5), and undesired activity at DAT was ablated.
Collapse
Affiliation(s)
- Carson
W. Reed
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jacob J. Kalbfleisch
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Madison J. Wong
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jordan P. Washecheck
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Ashton Hunter
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Alice L. Rodriguez
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Anna L. Blobaum
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt
Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Colleen M. Niswender
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt
Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
36
|
Xiang Y, Tanaka Y, Patterson B, Hwang SM, Hysolli E, Cakir B, Kim KY, Wang W, Kang YJ, Clement EM, Zhong M, Lee SH, Cho YS, Patra P, Sullivan GJ, Weissman SM, Park IH. Dysregulation of BRD4 Function Underlies the Functional Abnormalities of MeCP2 Mutant Neurons. Mol Cell 2020; 79:84-98.e9. [PMID: 32526163 PMCID: PMC7375197 DOI: 10.1016/j.molcel.2020.05.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 03/04/2020] [Accepted: 05/12/2020] [Indexed: 12/22/2022]
Abstract
Rett syndrome (RTT), mainly caused by mutations in methyl-CpG binding protein 2 (MeCP2), is one of the most prevalent intellectual disorders without effective therapies. Here, we used 2D and 3D human brain cultures to investigate MeCP2 function. We found that MeCP2 mutations cause severe abnormalities in human interneurons (INs). Surprisingly, treatment with a BET inhibitor, JQ1, rescued the molecular and functional phenotypes of MeCP2 mutant INs. We uncovered that abnormal increases in chromatin binding of BRD4 and enhancer-promoter interactions underlie the abnormal transcription in MeCP2 mutant INs, which were recovered to normal levels by JQ1. We revealed cell-type-specific transcriptome impairment in MeCP2 mutant region-specific human brain organoids that were rescued by JQ1. Finally, JQ1 ameliorated RTT-like phenotypes in mice. These data demonstrate that BRD4 dysregulation is a critical driver for RTT etiology and suggest that targeting BRD4 could be a potential therapeutic opportunity for RTT.
Collapse
Affiliation(s)
- Yangfei Xiang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yoshiaki Tanaka
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Benjamin Patterson
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Sung-Min Hwang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Eriona Hysolli
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Bilal Cakir
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kun-Yong Kim
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Wanshan Wang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Young-Jin Kang
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ethan M Clement
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Mei Zhong
- Department of Cell Biology, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Sang-Hun Lee
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yee Sook Cho
- Regenerative Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Republic of Korea
| | - Prabir Patra
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA; Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA
| | - Gareth J Sullivan
- Department of Molecular Medicine, Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, Oslo University Hospital and University of Oslo, Oslo 0424, Norway; Department of Pediatric Research, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
| | - Sherman M Weissman
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
37
|
Khoury ES, Sharma A, Ramireddy RR, Thomas AG, Alt J, Fowler A, Rais R, Tsukamoto T, Blue ME, Slusher B, Kannan S, Kannan RM. Dendrimer-conjugated glutaminase inhibitor selectively targets microglial glutaminase in a mouse model of Rett syndrome. Am J Cancer Res 2020; 10:5736-5748. [PMID: 32483415 PMCID: PMC7254984 DOI: 10.7150/thno.41714] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Elevated glutamate production and release from glial cells is a common feature of many CNS disorders. Inhibitors of glutaminase (GLS), the enzyme responsible for converting glutamine to glutamate have been developed to target glutamate overproduction. However, many GLS inhibitors have poor aqueous solubility, are unable to cross the blood brain barrier, or demonstrate significant toxicity when given systemically, precluding translation. Enhanced aqueous solubility and systemic therapy targeted to activated glia may address this challenge. Here we examine the impact of microglial-targeted GLS inhibition in a mouse model of Rett syndrome (RTT), a developmental disorder with no viable therapies, manifesting profound central nervous system effects, in which elevated glutamatergic tone, upregulation of microglial GLS, oxidative stress and neuroimmune dysregulation are key features. Methods: To enable this, we conjugated a potent glutaminase inhibitor, N-(5-{2-[2-(5-amino-[1,3,4]thiadiazol-2-yl)-ethylsulfanyl]-ethyl}-[1,3,4]thiadiazol-2-yl)-2-phenyl-acetamide (JHU29) to a generation 4 hydroxyl PAMAM dendrimer (D-JHU29). We then examined the effect of D-JHU29 in organotypic slice culture on glutamate release. We also examined GLS activity in microglial and non-microglial cells, and neurobehavioral phenotype after systemic administration of D-JHU29 in a mouse model of RTT. Results: We report successful conjugation of JHU29 to dendrimer resulting in enhanced water solubility compared to free JHU29. D-JHU29 reduced the excessive glutamate release observed in tissue culture slices in a clinically relevant Mecp2-knockout (KO) RTT mouse. Microglia isolated from Mecp2-KO mice demonstrated upregulation of GLS activity that normalized to wild-type levels following systemic treatment with D-JHU29. Neurobehavioral assessments in D-JHU29 treated Mecp2-KO mice revealed selective improvements in mobility. Conclusion: These findings demonstrate that glutaminase inhibitors conjugated to dendrimers are a viable mechanism to selectively inhibit microglial GLS to reduce glutamate production and improve mobility in a mouse model of RTT, with broader implications for selectively targeting this pathway in other neurodegenerative disorders.
Collapse
|
38
|
Fisher NM, Gould RW, Gogliotti RG, McDonald AJ, Badivuku H, Chennareddy S, Buch AB, Moore AM, Jenkins MT, Robb WH, Lindsley CW, Jones CK, Conn PJ, Niswender CM. Phenotypic profiling of mGlu 7 knockout mice reveals new implications for neurodevelopmental disorders. GENES BRAIN AND BEHAVIOR 2020; 19:e12654. [PMID: 32248644 DOI: 10.1111/gbb.12654] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/03/2020] [Accepted: 03/26/2020] [Indexed: 12/17/2022]
Abstract
Neurodevelopmental disorders are characterized by deficits in communication, cognition, attention, social behavior and/or motor control. Previous studies have pointed to the involvement of genes that regulate synaptic structure and function in the pathogenesis of these disorders. One such gene, GRM7, encodes the metabotropic glutamate receptor 7 (mGlu7 ), a G protein-coupled receptor that regulates presynaptic neurotransmitter release. Mutations and polymorphisms in GRM7 have been associated with neurodevelopmental disorders in clinical populations; however, limited preclinical studies have evaluated mGlu7 in the context of this specific disease class. Here, we show that the absence of mGlu7 in mice is sufficient to alter phenotypes within the domains of social behavior, associative learning, motor function, epilepsy and sleep. Moreover, Grm7 knockout mice exhibit an attenuated response to amphetamine. These findings provide rationale for further investigation of mGlu7 as a potential therapeutic target for neurodevelopmental disorders such as idiopathic autism, attention deficit hyperactivity disorder and Rett syndrome.
Collapse
Affiliation(s)
- Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Robert W Gould
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Rocco G Gogliotti
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Annalise J McDonald
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Hana Badivuku
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Susmita Chennareddy
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Aditi B Buch
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Annah M Moore
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Matthew T Jenkins
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - W Hudson Robb
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
39
|
Age-dependent impairment of metabotropic glutamate receptor 2-dependent long-term depression in the mouse striatum by chronic ethanol exposure. Alcohol 2020; 82:11-21. [PMID: 31233806 PMCID: PMC6925350 DOI: 10.1016/j.alcohol.2019.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/22/2022]
Abstract
Chronic alcohol exposure is associated with increased reliance on behavioral strategies involving the dorsolateral striatum (DLS), including habitual or stimulus-response behaviors. Presynaptic G protein-coupled receptors (GPCRs) on cortical and thalamic inputs to the DLS inhibit glutamate release, and alcohol-induced disruption of presynaptic GPCR function represents a mechanism by which alcohol could disinhibit DLS neurons and thus bias toward use of DLS-dependent behaviors. Metabotropic glutamate receptor 2 (mGlu2) is a Gi/o-coupled GPCR that robustly modulates glutamate transmission in the DLS, inducing long-term depression (LTD) at both cortical and thalamic synapses. Loss of mGlu2 function has recently been associated with increased ethanol seeking and consumption, but the ability of alcohol to produce adaptations in mGlu2 function in the DLS has not been investigated. We exposed male C57Bl/6J mice to a 2-week chronic intermittent ethanol (CIE) paradigm followed by a brief withdrawal period, then used whole-cell patch clamp recordings of glutamatergic transmission in the striatum to assess CIE effects on mGlu2-mediated synaptic plasticity. We report that CIE differentially disrupts mGlu2-mediated long-term depression in the DLS vs. dorsomedial striatum (DMS). Interestingly, CIE-induced impairment of mGlu2-LTD in the dorsolateral striatum is only observed when alcohol exposure occurs during adolescence. Incubation of striatal slices from CIE-exposed adolescent mice with a positive allosteric modulator of mGlu2 fully rescues mGlu2-LTD. In contrast to the 2-week CIE paradigm, acute exposure of striatal slices to ethanol concentrations that mimic ethanol levels during CIE exposure fails to disrupt mGlu2-LTD. We did not observe a reduction of mGlu2 mRNA or protein levels following CIE exposure, suggesting that alcohol effects on mGlu2 occur at the functional level. Our findings contribute to growing evidence that adolescents are uniquely vulnerable to certain alcohol-induced neuroadaptations, and identify enhancement of mGlu2 activity as a strategy to reverse the effects of adolescent alcohol exposure on DLS physiology.
Collapse
|
40
|
Yousefzadeh SA, Hesslow G, Shumyatsky GP, Meck WH. Internal Clocks, mGluR7 and Microtubules: A Primer for the Molecular Encoding of Target Durations in Cerebellar Purkinje Cells and Striatal Medium Spiny Neurons. Front Mol Neurosci 2020; 12:321. [PMID: 31998074 PMCID: PMC6965020 DOI: 10.3389/fnmol.2019.00321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 12/16/2019] [Indexed: 12/16/2022] Open
Abstract
The majority of studies in the field of timing and time perception have generally focused on sub- and supra-second time scales, specific behavioral processes, and/or discrete neuronal circuits. In an attempt to find common elements of interval timing from a broader perspective, we review the literature and highlight the need for cell and molecular studies that can delineate the neural mechanisms underlying temporal processing. Moreover, given the recent attention to the function of microtubule proteins and their potential contributions to learning and memory consolidation/re-consolidation, we propose that these proteins play key roles in coding temporal information in cerebellar Purkinje cells (PCs) and striatal medium spiny neurons (MSNs). The presence of microtubules at relevant neuronal sites, as well as their adaptability, dynamic structure, and longevity, makes them a suitable candidate for neural plasticity at both intra- and inter-cellular levels. As a consequence, microtubules appear capable of maintaining a temporal code or engram and thereby regulate the firing patterns of PCs and MSNs known to be involved in interval timing. This proposed mechanism would control the storage of temporal information triggered by postsynaptic activation of mGluR7. This, in turn, leads to alterations in microtubule dynamics through a "read-write" memory process involving alterations in microtubule dynamics and their hexagonal lattice structures involved in the molecular basis of temporal memory.
Collapse
Affiliation(s)
- S. Aryana Yousefzadeh
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| | - Germund Hesslow
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Gleb P. Shumyatsky
- Department of Genetics, Rutgers University, Piscataway, NJ, United States
| | - Warren H. Meck
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| |
Collapse
|
41
|
Ribeiro MC, MacDonald JL. Sex differences in Mecp2-mutant Rett syndrome model mice and the impact of cellular mosaicism in phenotype development. Brain Res 2020; 1729:146644. [PMID: 31904347 DOI: 10.1016/j.brainres.2019.146644] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/08/2019] [Accepted: 12/31/2019] [Indexed: 12/29/2022]
Abstract
There is currently no effective treatment for Rett syndrome (RTT), a severe X-linked progressive neurodevelopmental disorder caused by mutations in the transcriptional regulator MECP2. Because MECP2 is subjected to X-inactivation, most affected individuals are female heterozygotes who display cellular mosaicism for normal and mutant MECP2. Males who are hemizygous for mutant MECP2 are more severely affected than heterozygous females and rarely survive. Mecp2 loss-of-function is less severe in mice, however, and male hemizygous null mice not only survive until adulthood, they have been the most commonly studied model system. Although heterozygous female mice better recapitulate human RTT, they have not been as thoroughly characterized. This is likely because of the added experimental challenges that they present, including delayed and more variable phenotypic progression and cellular mosaicism due to X-inactivation. In this review, we compare phenotypes of Mecp2 heterozygous female mice and male hemizygous null mouse models. Further, we discuss the complexities that arise from the many cell-type and tissue-type specific roles of MeCP2, as well as the combination of cell-autonomous and non-cell-autonomous disruptions that result from Mecp2 loss-of-function. This is of particular importance in the context of the female heterozygous brain, composed of a mixture of MeCP2+ and MeCP2- cells, the ratio of which can alter RTT phenotypes in the case of skewed X-inactivation. The goal of this review is to provide a clearer understanding of the pathophysiological differences between the mouse models, which is an essential consideration in the design of future pre-clinical studies.
Collapse
Affiliation(s)
- Mayara C Ribeiro
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States
| | - Jessica L MacDonald
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States.
| |
Collapse
|
42
|
ELFN2 is a postsynaptic cell adhesion molecule with essential roles in controlling group III mGluRs in the brain and neuropsychiatric behavior. Mol Psychiatry 2019; 24:1902-1919. [PMID: 31485013 PMCID: PMC6874751 DOI: 10.1038/s41380-019-0512-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 07/17/2019] [Accepted: 07/26/2019] [Indexed: 12/17/2022]
Abstract
The functional characterization of the GPCR interactome has predominantly focused on intracellular binding partners; however, the recent emergence of transsynaptic GPCR complexes represents an additional dimension to GPCR function that has previously been unaccounted for in drug discovery. Here, we characterize ELFN2 as a novel postsynaptic adhesion molecule with a distinct expression pattern throughout the brain and a selective binding with group III metabotropic glutamate receptors (mGluRs) in trans. Using a transcellular GPCR signaling platform, we report that ELFN2 critically alters group III mGluR secondary messenger signaling by directly altering G protein coupling kinetics and efficacy. Loss of ELFN2 in mice results in the selective downregulation of group III mGluRs and dysregulated glutamatergic synaptic transmission. Elfn2 knockout (Elfn2 KO) mice also feature a range of neuropsychiatric manifestations including seizure susceptibility, hyperactivity, and anxiety/compulsivity, which can be rescued by pharmacological augmentation of group III mGluRs. Thus, we conclude that extracellular transsynaptic scaffolding by ELFN2 in the brain is a cardinal organizational feature of group III mGluRs essential for their signaling properties and brain function.
Collapse
|
43
|
Smith ES, Smith DR, Eyring C, Braileanu M, Smith-Connor KS, Ei Tan Y, Fowler AY, Hoffman GE, Johnston MV, Kannan S, Blue ME. Altered trajectories of neurodevelopment and behavior in mouse models of Rett syndrome. Neurobiol Learn Mem 2019; 165:106962. [PMID: 30502397 PMCID: PMC8040058 DOI: 10.1016/j.nlm.2018.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 10/17/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022]
Abstract
Rett Syndrome (RTT) is a genetic disorder that is caused by mutations in the x-linked gene coding for methyl-CpG-biding-protein 2 (MECP2) and that mainly affects females. Male and female transgenic mouse models of RTT have been studied extensively, and we have learned a great deal regarding RTT neuropathology and how MeCP2 deficiency may be influencing brain function and maturation. In this manuscript we review what is known concerning structural and coinciding functional and behavioral deficits in RTT and in mouse models of MeCP2 deficiency. We also introduce our own corroborating data regarding behavioral phenotype and morphological alterations in volume of the cortex and striatum and the density of neurons, aberrations in experience-dependent plasticity within the barrel cortex and the impact of MeCP2 loss on glial structure. We conclude that regional structural changes in genetic models of RTT show great similarity to the alterations in brain structure of patients with RTT. These region-specific modifications often coincide with phenotype onset and contribute to larger issues of circuit connectivity, progression, and severity. Although the alterations seen in mouse models of RTT appear to be primarily due to cell-autonomous effects, there are also non-cell autonomous mechanisms including those caused by MeCP2-deficient glia that negatively impact healthy neuronal function. Collectively, this body of work has provided a solid foundation on which to continue to build our understanding of the role of MeCP2 on neuronal and glial structure and function, its greater impact on neural development, and potential new therapeutic avenues.
Collapse
Affiliation(s)
- Elizabeth S Smith
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dani R Smith
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Charlotte Eyring
- The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Maria Braileanu
- Undergraduate Program in Neuroscience, The Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Karen S Smith-Connor
- The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Yew Ei Tan
- Perdana University Graduate School of Medicine, Kuala Lumpur, Malaysia
| | - Amanda Y Fowler
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA
| | - Gloria E Hoffman
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA
| | - Michael V Johnston
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Mary E Blue
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA.
| |
Collapse
|
44
|
Early Postnatal Treatment with Valproate Induces Gad1 Promoter Remodeling in the Brain and Reduces Apnea Episodes in Mecp2-Null Mice. Int J Mol Sci 2019; 20:ijms20205177. [PMID: 31635390 PMCID: PMC6834123 DOI: 10.3390/ijms20205177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 11/17/2022] Open
Abstract
The deletion of Mecp2, the gene encoding methyl-CpG-binding protein 2, causes severe breathing defects and developmental anomalies in mammals. In Mecp2-null mice, impaired GABAergic neurotransmission is demonstrated at the early stage of life. GABAergic dysfunction in neurons in the rostral ventrolateral medulla (RVLM) is considered as a primary cause of breathing abnormality in Mecp2-null mice, but its molecular mechanism is unclear. Here, we report that mRNA expression levels of Gad1, which encodes glutamate decarboxylase 67 (GAD67), in the RVLM of Mecp2-null (Mecp2-/y, B6.129P2(C)-Mecp2tm1.1Bird/J) mice is closely related to the methylation status of its promoter, and valproate (VPA) can upregulate transcription from Gad1 through epigenetic mechanisms. The administration of VPA (300 mg/kg/day) together with L-carnitine (30 mg/kg/day) from day 8 to day 14 after birth increased Gad1 mRNA expression in the RVLM and reduced apnea counts in Mecp2-/y mice on postnatal day 15. Cytosine methylation levels in the Gad1 promoter were higher in the RVLM of Mecp2-/y mice compared to wild-type mice born to C57BL/6J females, while VPA treatment decreased the methylation levels in Mecp2-/y mice. Chromatin immunoprecipitation assay revealed that the VPA treatment reduced the binding of methyl-CpG binding domain protein 1 (MBD1) to the Gad1 promoter in Mecp2-/y mice. These results suggest that VPA improves breathing of Mecp2-/y mice by reducing the Gad1 promoter methylation, which potentially leads to the enhancement of GABAergic neurotransmission in the RVLM.
Collapse
|
45
|
Kadam SD, Sullivan BJ, Goyal A, Blue ME, Smith-Hicks C. Rett Syndrome and CDKL5 Deficiency Disorder: From Bench to Clinic. Int J Mol Sci 2019; 20:ijms20205098. [PMID: 31618813 PMCID: PMC6834180 DOI: 10.3390/ijms20205098] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/18/2022] Open
Abstract
Rett syndrome (RTT) and CDKL5 deficiency disorder (CDD) are two rare X-linked developmental brain disorders with overlapping but distinct phenotypic features. This review examines the impact of loss of methyl-CpG-binding protein 2 (MeCP2) and cyclin-dependent kinase-like 5 (CDKL5) on clinical phenotype, deficits in synaptic- and circuit-homeostatic mechanisms, seizures, and sleep. In particular, we compare the overlapping and contrasting features between RTT and CDD in clinic and in preclinical studies. Finally, we discuss lessons learned from recent clinical trials while reviewing the findings from pre-clinical studies.
Collapse
Affiliation(s)
- Shilpa D Kadam
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Brennan J Sullivan
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
| | - Archita Goyal
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
| | - Mary E Blue
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Constance Smith-Hicks
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
46
|
Surveying heterocycles as amide bioisosteres within a series of mGlu7 NAMs: Discovery of VU6019278. Bioorg Med Chem Lett 2019; 29:1211-1214. [DOI: 10.1016/j.bmcl.2019.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/18/2019] [Accepted: 03/13/2019] [Indexed: 12/19/2022]
|
47
|
Gogliotti RG, Niswender CM. A Coordinated Attack: Rett Syndrome Therapeutic Development. Trends Pharmacol Sci 2019; 40:233-236. [PMID: 30905360 DOI: 10.1016/j.tips.2019.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/06/2019] [Accepted: 02/06/2019] [Indexed: 10/27/2022]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the Methyl CpG binding protein 2 (MeCP2) gene. This Science & Society article focuses on pharmacological strategies that attack RTT treatment from multiple angles, including drug repurposing and de novo discovery efforts, and discusses the impacts of preclinical study design and translationally relevant outcome measures.
Collapse
Affiliation(s)
- Rocco G Gogliotti
- Vanderbilt Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Colleen M Niswender
- Vanderbilt Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
48
|
Stansley BJ, Conn PJ. Neuropharmacological Insight from Allosteric Modulation of mGlu Receptors. Trends Pharmacol Sci 2019; 40:240-252. [PMID: 30824180 PMCID: PMC6445545 DOI: 10.1016/j.tips.2019.02.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/06/2019] [Accepted: 02/06/2019] [Indexed: 12/11/2022]
Abstract
The metabotropic glutamate (mGlu) receptors are a family of G-protein-coupled receptors (GPCRs) that regulate cell physiology throughout the nervous system. The potential of mGlu receptors as therapeutic targets has been bolstered by current research that has provided insight into the diverse modes of mGlu activation and signaling. In particular, the allosteric modulation of mGlu receptors represents a major area of focus in studies of basic pharmacology as well as drug development, largely due to the high subtype specificity achievable by targeting allosteric sites on mGlu receptors. These provide sophisticated regulation of neuronal excitability and synaptic transmission to influence behavioral output. Here, we review how these allosteric mechanisms have been leveraged preclinically to demonstrate the therapeutic potential of allosteric modulators for neurological and neuropsychiatric disorders, such as autism, cognitive impairment, Parkinson's disease (PD), stress, and schizophrenia.
Collapse
Affiliation(s)
- Branden J Stansley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
49
|
Treating Rett syndrome: from mouse models to human therapies. Mamm Genome 2019; 30:90-110. [PMID: 30820643 PMCID: PMC6606665 DOI: 10.1007/s00335-019-09793-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/09/2019] [Indexed: 02/06/2023]
Abstract
Rare diseases are very difficult to study mechanistically and to develop therapies for because of the scarcity of patients. Here, the rare neuro-metabolic disorder Rett syndrome (RTT) is discussed as a prototype for precision medicine, demonstrating how mouse models have led to an understanding of the development of symptoms. RTT is caused by mutations in the X-linked gene methyl-CpG-binding protein 2 (MECP2). Mecp2-mutant mice are being used in preclinical studies that target the MECP2 gene directly, or its downstream pathways. Importantly, this work may improve the health of RTT patients. Clinical presentation may vary widely among individuals based on their mutation, but also because of the degree of X chromosome inactivation and the presence of modifier genes. Because it is a complex disorder involving many organ systems, it is likely that recovery of RTT patients will involve a combination of treatments. Precision medicine is warranted to provide the best efficacy to individually treat RTT patients.
Collapse
|
50
|
Reed CW, Yohn SE, Washecheck JP, Roenfanz HF, Quitalig MC, Luscombe VB, Jenkins MT, Rodriguez AL, Engers DW, Blobaum AL, Conn PJ, Niswender CM, Lindsley CW. Discovery of an Orally Bioavailable and Central Nervous System (CNS) Penetrant mGlu 7 Negative Allosteric Modulator (NAM) in Vivo Tool Compound: N-(2-(1 H-1,2,4-triazol-1-yl)-5-(trifluoromethoxy)phenyl)-4-(cyclopropylmethoxy)-3-methoxybenzamide (VU6012962). J Med Chem 2019; 62:1690-1695. [PMID: 30608678 DOI: 10.1021/acs.jmedchem.8b01810] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Herein, we report the discovery of a new, orally bioavailable and CNS-penetrant metabotropic glutamate receptor 7 (mGlu7) negative allosteric modulator (NAM) that achieves exposure in cerebral spinal fluid (CSF) 2.5× above the in vitro IC50 at minimum effective doses (MEDs) of 3 mg/kg in preclinical anxiety models.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - P Jeffrey Conn
- Vanderbilt Kennedy Center , Vanderbilt University School of Medicine , Nashville , Tennessee 37232 , United States
| | - Colleen M Niswender
- Vanderbilt Kennedy Center , Vanderbilt University School of Medicine , Nashville , Tennessee 37232 , United States
| | | |
Collapse
|