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
|
Ferranti AS, Luessen DJ, Niswender CM. Novel pharmacological targets for GABAergic dysfunction in ADHD. Neuropharmacology 2024; 249:109897. [PMID: 38462041 DOI: 10.1016/j.neuropharm.2024.109897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
Attention deficit/hyperactivity disorder (ADHD) is a neurodevelopment disorder that affects approximately 5% of the population. The disorder is characterized by impulsivity, hyperactivity, and deficits in attention and cognition, although symptoms vary across patients due to the heterogenous and polygenic nature of the disorder. Stimulant medications are the standard of care treatment for ADHD patients, and their effectiveness has led to the dopaminergic hypothesis of ADHD in which deficits in dopaminergic signaling, especially in cortical brain regions, mechanistically underly ADHD pathophysiology. Despite their effectiveness in many individuals, almost one-third of patients do not respond to stimulant treatments and the long-term negative side effects of these medications remain unclear. Emerging clinical evidence is beginning to highlight an important role of dysregulated excitatory/inhibitory (E/I) balance in ADHD. These deficits in E/I balance are related to functional abnormalities in glutamate and Gamma-Aminobutyric Acid (GABA) signaling in the brain, with increasing emphasis placed on GABAergic interneurons driving specific aspects of ADHD pathophysiology. Recent genome-wide association studies (GWAS) have also highlighted how genes associated with GABA function are mutated in human populations with ADHD, resulting in the generation of several new genetic mouse models of ADHD. This review will discuss how GABAergic dysfunction underlies ADHD pathophysiology, and how specific receptors/proteins related to GABAergic interneuron dysfunction may be pharmacologically targeted to treat ADHD in subpopulations with specific comorbidities and symptom domains. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Anthony S Ferranti
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
| | - Deborah J Luessen
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Lukacs IP, Francavilla R, Field M, Hunter E, Howarth M, Horie S, Plaha P, Stacey R, Livermore L, Ansorge O, Tamas G, Somogyi P. Differential effects of group III metabotropic glutamate receptors on spontaneous inhibitory synaptic currents in spine-innervating double bouquet and parvalbumin-expressing dendrite-targeting GABAergic interneurons in human neocortex. Cereb Cortex 2023; 33:2101-2142. [PMID: 35667019 PMCID: PMC9977385 DOI: 10.1093/cercor/bhac195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/12/2022] Open
Abstract
Diverse neocortical GABAergic neurons specialize in synaptic targeting and their effects are modulated by presynaptic metabotropic glutamate receptors (mGluRs) suppressing neurotransmitter release in rodents, but their effects in human neocortex are unknown. We tested whether activation of group III mGluRs by L-AP4 changes GABAA receptor-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) in 2 distinct dendritic spine-innervating GABAergic interneurons recorded in vitro in human neocortex. Calbindin-positive double bouquet cells (DBCs) had columnar "horsetail" axons descending through layers II-V innervating dendritic spines (48%) and shafts, but not somata of pyramidal and nonpyramidal neurons. Parvalbumin-expressing dendrite-targeting cell (PV-DTC) axons extended in all directions innervating dendritic spines (22%), shafts (65%), and somata (13%). As measured, 20% of GABAergic neuropil synapses innervate spines, hence DBCs, but not PV-DTCs, preferentially select spine targets. Group III mGluR activation paradoxically increased the frequency of sIPSCs in DBCs (to median 137% of baseline) but suppressed it in PV-DTCs (median 92%), leaving the amplitude unchanged. The facilitation of sIPSCs in DBCs may result from their unique GABAergic input being disinhibited via network effect. We conclude that dendritic spines receive specialized, diverse GABAergic inputs, and group III mGluRs differentially regulate GABAergic synaptic transmission to distinct GABAergic cell types in human cortex.
Collapse
Affiliation(s)
- Istvan P Lukacs
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | | | - Martin Field
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Emily Hunter
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Michael Howarth
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Sawa Horie
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Puneet Plaha
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Richard Stacey
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Laurent Livermore
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Gabor Tamas
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, 6726 Szeged, Hungary
| | - Peter Somogyi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
6
|
Holter KM, Pierce BE, Gould RW. Metabotropic glutamate receptor function and regulation of sleep-wake cycles. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:93-175. [PMID: 36868636 PMCID: PMC10973983 DOI: 10.1016/bs.irn.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Metabotropic glutamate (mGlu) receptors are the most abundant family of G-protein coupled receptors and are widely expressed throughout the central nervous system (CNS). Alterations in glutamate homeostasis, including dysregulations in mGlu receptor function, have been indicated as key contributors to multiple CNS disorders. Fluctuations in mGlu receptor expression and function also occur across diurnal sleep-wake cycles. Sleep disturbances including insomnia are frequently comorbid with neuropsychiatric, neurodevelopmental, and neurodegenerative conditions. These often precede behavioral symptoms and/or correlate with symptom severity and relapse. Chronic sleep disturbances may also be a consequence of primary symptom progression and can exacerbate neurodegeneration in disorders including Alzheimer's disease (AD). Thus, there is a bidirectional relationship between sleep disturbances and CNS disorders; disrupted sleep may serve as both a cause and a consequence of the disorder. Importantly, comorbid sleep disturbances are rarely a direct target of primary pharmacological treatments for neuropsychiatric disorders even though improving sleep can positively impact other symptom clusters. This chapter details known roles of mGlu receptor subtypes in both sleep-wake regulation and CNS disorders focusing on schizophrenia, major depressive disorder, post-traumatic stress disorder, AD, and substance use disorder (cocaine and opioid). In this chapter, preclinical electrophysiological, genetic, and pharmacological studies are described, and, when possible, human genetic, imaging, and post-mortem studies are also discussed. In addition to reviewing the important relationships between sleep, mGlu receptors, and CNS disorders, this chapter highlights the development of selective mGlu receptor ligands that hold promise for improving both primary symptoms and sleep disturbances.
Collapse
Affiliation(s)
- Kimberly M Holter
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Bethany E Pierce
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Robert W Gould
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
7
|
Reed CW, Rodriguez AL, Kalbfleisch JJ, Seto M, Jenkins MT, Blobaum AL, Chang S, Lindsley CW, Niswender CM. Development and profiling of mGlu 7 NAMs with a range of saturable inhibition of agonist responses in vitro. Bioorg Med Chem Lett 2022; 74:128923. [PMID: 35944850 PMCID: PMC10015594 DOI: 10.1016/j.bmcl.2022.128923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 11/02/2022]
Abstract
We describe here a series of metabotropic glutamate receptor 7 (mGlu7) negative allosteric modulators (NAMs) with a saturable range of activity in inhibiting responses to an orthosteric agonist in two distinct in vitro pharmacological assays. The range of inhibition among compounds in this scaffold provides highly structurally related ligands with differential degrees of receptor blockade that can be used to understand inhibitory efficacy profiles in native tissue or in vivo.
Collapse
Affiliation(s)
- Carson W Reed
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Alice L Rodriguez
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Jacob J Kalbfleisch
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Mabel Seto
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Matthew T Jenkins
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Anna L Blobaum
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Sichen Chang
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Vanderbilt Department of Chemistry, Vanderbilt University, Nashville, TN 37232, United States
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, United States.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Bauminger H, Gaisler-Salomon I. Beyond NMDA Receptors: Homeostasis at the Glutamate Tripartite Synapse and Its Contributions to Cognitive Dysfunction in Schizophrenia. Int J Mol Sci 2022; 23:8617. [PMID: 35955750 PMCID: PMC9368772 DOI: 10.3390/ijms23158617] [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] [Received: 06/28/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 02/01/2023] Open
Abstract
Cognitive deficits are core symptoms of schizophrenia but remain poorly addressed by dopamine-based antipsychotic medications. Glutamate abnormalities are implicated in schizophrenia-related cognitive deficits. While the role of the NMDA receptor has been extensively studied, less attention was given to other components that control glutamate homeostasis. Glutamate dynamics at the tripartite synapse include presynaptic and postsynaptic components and are tightly regulated by neuron-astrocyte crosstalk. Here, we delineate the role of glutamate homeostasis at the tripartite synapse in schizophrenia-related cognitive dysfunction. We focus on cognitive domains that can be readily measured in humans and rodents, i.e., working memory, recognition memory, cognitive flexibility, and response inhibition. We describe tasks used to measure cognitive function in these domains in humans and rodents, and the relevance of glutamate alterations in these domains. Next, we delve into glutamate tripartite synaptic components and summarize findings that implicate the relevance of these components to specific cognitive domains. These collective findings indicate that neuron-astrocyte crosstalk at the tripartite synapse is essential for cognition, and that pre- and postsynaptic components play a critical role in maintaining glutamate homeostasis and cognitive well-being. The contribution of these components to cognitive function should be considered in order to better understand the role played by glutamate signaling in cognition and develop efficient pharmacological treatment avenues for schizophrenia treatment-resistant symptoms.
Collapse
Affiliation(s)
- Hagar Bauminger
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Inna Gaisler-Salomon
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
11
|
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
|
12
|
Luessen DJ, Conn PJ. Allosteric Modulators of Metabotropic Glutamate Receptors as Novel Therapeutics for Neuropsychiatric Disease. Pharmacol Rev 2022; 74:630-661. [PMID: 35710132 PMCID: PMC9553119 DOI: 10.1124/pharmrev.121.000540] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors, a family of G-protein-coupled receptors, have been identified as novel therapeutic targets based on extensive research supporting their diverse contributions to cell signaling and physiology throughout the nervous system and important roles in regulating complex behaviors, such as cognition, reward, and movement. Thus, targeting mGlu receptors may be a promising strategy for the treatment of several brain disorders. Ongoing advances in the discovery of subtype-selective allosteric modulators for mGlu receptors has provided an unprecedented opportunity for highly specific modulation of signaling by individual mGlu receptor subtypes in the brain by targeting sites distinct from orthosteric or endogenous ligand binding sites on mGlu receptors. These pharmacological agents provide the unparalleled opportunity to selectively regulate neuronal excitability, synaptic transmission, and subsequent behavioral output pertinent to many brain disorders. Here, we review preclinical and clinical evidence supporting the utility of mGlu receptor allosteric modulators as novel therapeutic approaches to treat neuropsychiatric diseases, such as schizophrenia, substance use disorders, and stress-related disorders.
Collapse
|
13
|
Bodzęta A, Berger F, MacGillavry HD. Subsynaptic mobility of presynaptic mGluR types is differentially regulated by intra- and extracellular interactions. Mol Biol Cell 2022; 33:ar66. [PMID: 35511883 PMCID: PMC9635276 DOI: 10.1091/mbc.e21-10-0484] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Presynaptic metabotropic glutamate receptors (mGluRs) are essential for the control of synaptic transmission. However, how the subsynaptic dynamics of these receptors is controlled and contributes to synaptic signaling remain poorly understood quantitatively. Particularly, since the affinity of individual mGluR subtypes for glutamate differs considerably, the activation of mGluR subtypes critically depends on their precise subsynaptic distribution. Here, using superresolution microscopy and single-molecule tracking, we unravel novel molecular mechanisms that control the nanoscale distribution and mobility of presynaptic mGluRs in hippocampal neurons. We demonstrate that the high-affinity group II receptor mGluR2 localizes diffusely along the axon, and is highly mobile, while the low-affinity group III receptor mGluR7 is stably anchored at the active zone. We demonstrate that intracellular interactions modulate surface diffusion of mGluR2, while immobilization of mGluR7 at the active zone relies on its extracellular domain. Receptor activation or increases in synaptic activity do not alter the surface mobility of presynaptic mGluRs. Finally, computational modeling of presynaptic mGluR activity revealed that this particular nanoscale arrangement directly impacts their ability to modulate neurotransmitter release. Altogether, this study demonstrates that distinct mechanisms control surface mobility of presynaptic mGluRs to contribute differentially to glutamatergic synaptic transmission.
Collapse
Affiliation(s)
- Anna Bodzęta
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, The Netherlands
| | - Florian Berger
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, The Netherlands
| | - Harold D MacGillavry
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, The Netherlands
| |
Collapse
|
14
|
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
|
15
|
Membrane trafficking and positioning of mGluRs at presynaptic and postsynaptic sites of excitatory synapses. Neuropharmacology 2021; 200:108799. [PMID: 34592242 DOI: 10.1016/j.neuropharm.2021.108799] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 01/21/2023]
Abstract
The plethora of functions of glutamate in the brain are mediated by the complementary actions of ionotropic and metabotropic glutamate receptors (mGluRs). The ionotropic glutamate receptors carry most of the fast excitatory transmission, while mGluRs modulate transmission on longer timescales by triggering multiple intracellular signaling pathways. As such, mGluRs mediate critical aspects of synaptic transmission and plasticity. Interestingly, at synapses, mGluRs operate at both sides of the cleft, and thus bidirectionally exert the effects of glutamate. At postsynaptic sites, group I mGluRs act to modulate excitability and plasticity. At presynaptic sites, group II and III mGluRs act as auto-receptors, modulating release properties in an activity-dependent manner. Thus, synaptic mGluRs are essential signal integrators that functionally couple presynaptic and postsynaptic mechanisms of transmission and plasticity. Understanding how these receptors reach the membrane and are positioned relative to the presynaptic glutamate release site are therefore important aspects of synapse biology. In this review, we will discuss the currently known mechanisms underlying the trafficking and positioning of mGluRs at and around synapses, and how these mechanisms contribute to synaptic functioning. We will highlight outstanding questions and present an outlook on how recent technological developments will move this exciting research field forward.
Collapse
|
16
|
Woo MS, Ufer F, Rothammer N, Di Liberto G, Binkle L, Haferkamp U, Sonner JK, Engler JB, Hornig S, Bauer S, Wagner I, Egervari K, Raber J, Duvoisin RM, Pless O, Merkler D, Friese MA. Neuronal metabotropic glutamate receptor 8 protects against neurodegeneration in CNS inflammation. J Exp Med 2021; 218:e20201290. [PMID: 33661276 PMCID: PMC7938362 DOI: 10.1084/jem.20201290] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 12/17/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system with continuous neuronal loss. Treatment of clinical progression remains challenging due to lack of insights into inflammation-induced neurodegenerative pathways. Here, we show that an imbalance in the neuronal receptor interactome is driving glutamate excitotoxicity in neurons of MS patients and identify the MS risk-associated metabotropic glutamate receptor 8 (GRM8) as a decisive modulator. Mechanistically, GRM8 activation counteracted neuronal cAMP accumulation, thereby directly desensitizing the inositol 1,4,5-trisphosphate receptor (IP3R). This profoundly limited glutamate-induced calcium release from the endoplasmic reticulum and subsequent cell death. Notably, we found Grm8-deficient neurons to be more prone to glutamate excitotoxicity, whereas pharmacological activation of GRM8 augmented neuroprotection in mouse and human neurons as well as in a preclinical mouse model of MS. Thus, we demonstrate that GRM8 conveys neuronal resilience to CNS inflammation and is a promising neuroprotective target with broad therapeutic implications.
Collapse
Affiliation(s)
- Marcel S. Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Ufer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Giovanni Di Liberto
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Lars Binkle
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology, Hamburg, Germany
| | - Jana K. Sonner
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Sönke Hornig
- Experimentelle Neuropädiatrie, Klinik für Kinder und Jugendmedizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Bauer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Kristof Egervari
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR
| | - Robert M. Duvoisin
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology, Hamburg, Germany
| | - Doron Merkler
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Manuel A. Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
17
|
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
|
18
|
Srivastava A, Das B, Yao AY, Yan R. Metabotropic Glutamate Receptors in Alzheimer's Disease Synaptic Dysfunction: Therapeutic Opportunities and Hope for the Future. J Alzheimers Dis 2020; 78:1345-1361. [PMID: 33325389 PMCID: PMC8439550 DOI: 10.3233/jad-201146] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the presence of neuritic plaques and neurofibrillary tangles. The impaired synaptic plasticity and dendritic loss at the synaptic level is an early event associated with the AD pathogenesis. The abnormal accumulation of soluble oligomeric amyloid-β (Aβ), the major toxic component in amyloid plaques, is viewed to trigger synaptic dysfunctions through binding to several presynaptic and postsynaptic partners and thus to disrupt synaptic transmission. Over time, the abnormalities in neural transmission will result in cognitive deficits, which are commonly manifested as memory loss in AD patients. Synaptic plasticity is regulated through glutamate transmission, which is mediated by various glutamate receptors. Here we review recent progresses in the study of metabotropic glutamate receptors (mGluRs) in AD cognition. We will discuss the role of mGluRs in synaptic plasticity and their modulation as a possible strategy for AD cognitive improvement.
Collapse
Affiliation(s)
- Akriti Srivastava
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Brati Das
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Annie Y. Yao
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
19
|
Impact of the metabotropic glutamate receptor7 (mGlu 7) allosteric agonist, AMN082, on fear learning and memory and anxiety-like behavior. Eur J Pharmacol 2019; 858:172512. [PMID: 31260653 DOI: 10.1016/j.ejphar.2019.172512] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/19/2019] [Accepted: 06/27/2019] [Indexed: 12/30/2022]
Abstract
The present study was conducted to evaluate the influence of AMN082, the metabotropic glutamate receptor subtype 7 (mGlu7) allosteric agonist on different stages of memory processes connected with fear conditioning in the passive avoidance (PA) learning task in mice and negative emotional state (anxiety-like) induced by ethanol- and morphine-withdrawal in the elevated plus maze (EPM) test in rats. To perform the PA test, AMN082 (1.25, 2.5 and 5 mg/kg, i. p.) was injected to interfere with (or inhibit) acquisition, consolidation, and retrieval processes. The retention latency in each group was recorded using a step-through passive avoidance task 24 h after training. In turn, in ethanol- and morphine-withdrawal rats, the influence of AMN082 on anxiety-like behavior was estimated in the EPM test 24 h- (ethanol) and 72- h (morphine) after the last dose of repeated drug administrations. In all experimental groups, AMN082 at the dose of 5 mg/kg significantly decreased the step-through latency of long-term memory in the PA task. These AMN082 effects were reversed by MMPIP (10 mg/kg), the antagonist of mGlu7 receptor. AMN082 (2.5 and 5 mg/kg) also decreased ethanol- and morphine withdrawal-induced anxiety-like behavior in the EPM test, and this AMN082 (5 mg/kg) effect was counteracted by MMPIP pretreatment. Taken together, the results show that mGlu7 is involved in fear learning to the context and anxiety-like state connected with unpleasant experiences after ethanol- and morphine withdrawal in rodents. However, it appears that functional dissociation exists between these two AMN082 effects.
Collapse
|
20
|
Wrackmeyer U, Kaldrack J, Jüttner R, Pannasch U, Gimber N, Freiberg F, Purfürst B, Kainmueller D, Schmitz D, Haucke V, Rathjen FG, Gotthardt M. The cell adhesion protein CAR is a negative regulator of synaptic transmission. Sci Rep 2019; 9:6768. [PMID: 31043663 PMCID: PMC6494904 DOI: 10.1038/s41598-019-43150-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 04/17/2019] [Indexed: 11/09/2022] Open
Abstract
The Coxsackievirus and adenovirus receptor (CAR) is essential for normal electrical conductance in the heart, but its role in the postnatal brain is largely unknown. Using brain specific CAR knockout mice (KO), we discovered an unexpected role of CAR in neuronal communication. This includes increased basic synaptic transmission at hippocampal Schaffer collaterals, resistance to fatigue, and enhanced long-term potentiation. Spontaneous neurotransmitter release and speed of endocytosis are increased in KOs, accompanied by increased expression of the exocytosis associated calcium sensor synaptotagmin 2. Using proximity proteomics and binding studies, we link CAR to the exocytosis machinery as it associates with syntenin and synaptobrevin/VAMP2 at the synapse. Increased synaptic function does not cause adverse effects in KO mice, as behavior and learning are unaffected. Thus, unlike the connexin-dependent suppression of atrioventricular conduction in the cardiac knockout, communication in the CAR deficient brain is improved, suggesting a role for CAR in presynaptic processes.
Collapse
Affiliation(s)
- Uta Wrackmeyer
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Joanna Kaldrack
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - René Jüttner
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany.,Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Ulrike Pannasch
- Neuroscience Research Center, Cluster of Excellence NeuroCure, Charité, 10117, Berlin, Germany
| | - Niclas Gimber
- Department of Molecular Pharmacology and Cell Biology, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Fabian Freiberg
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Bettina Purfürst
- Core Facility Electron Microscopy, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Dagmar Kainmueller
- Biomedical Image Analysis, Max Delbrück Center for Molecular Medicine and Berlin Institute of Health, 13125, Berlin, Germany
| | - Dietmar Schmitz
- Neuroscience Research Center, Cluster of Excellence NeuroCure, Charité, 10117, Berlin, Germany
| | - Volker Haucke
- Department of Molecular Pharmacology and Cell Biology, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Fritz G Rathjen
- Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Michael Gotthardt
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany.
| |
Collapse
|
21
|
Girard B, Tuduri P, Moreno MP, Sakkaki S, Barboux C, Bouschet T, Varrault A, Vitre J, McCort-Tranchepain I, Dairou J, Acher F, Fagni L, Marchi N, Perroy J, Bertaso F. The mGlu7 receptor provides protective effects against epileptogenesis and epileptic seizures. Neurobiol Dis 2019; 129:13-28. [PMID: 31051234 DOI: 10.1016/j.nbd.2019.04.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/28/2019] [Accepted: 04/29/2019] [Indexed: 01/26/2023] Open
Abstract
Finding new targets to control or reduce seizure activity is essential to improve the management of epileptic patients. We hypothesized that activation of the pre-synaptic and inhibitory metabotropic glutamate receptor type 7 (mGlu7) reduces spontaneous seizures. We tested LSP2-9166, a recently developed mGlu7/4 agonist with unprecedented potency on mGlu7 receptors, in two paradigms of epileptogenesis. In a model of chemically induced epileptogenesis (pentylenetetrazole systemic injection), LSP2-9166 induces an anti-epileptogenic effect rarely observed in preclinical studies. In particular, we found a bidirectional modulation of seizure progression by mGlu4 and mGlu7 receptors, the latter preventing kindling. In the intra-hippocampal injection of kainic acid mouse model that mimics the human mesial temporal lobe epilepsy, we found that LSP2-9166 reduces seizure frequency and hippocampal sclerosis. LSP2-9166 also acts as an anti-seizure drug on established seizures in both models tested. Specific modulation of the mGlu7 receptor could represent a novel approach to reduce pathological network remodeling.
Collapse
Affiliation(s)
- Benoit Girard
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | - Pola Tuduri
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | | | - Sophie Sakkaki
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | | | | | - Annie Varrault
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | - Jihane Vitre
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | | | | | | | - Laurent Fagni
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | - Nicola Marchi
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | - Julie Perroy
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | | |
Collapse
|
22
|
O’Connor RM, McCafferty CP, Bravo JA, Singewald N, Holmes A, Cryan JF. Increased amygdalar metabotropic glutamate receptor 7 mRNA in a genetic mouse model of impaired fear extinction. Psychopharmacology (Berl) 2019; 236:265-272. [PMID: 30215216 PMCID: PMC6739849 DOI: 10.1007/s00213-018-5031-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/05/2018] [Indexed: 12/18/2022]
Abstract
RATIONALE Post-traumatic stress disorder (PTSD) is a devastating anxiety-related disorder which develops subsequent to a severe psychologically traumatic event. Only ~ 9% of people who experience such a trauma develop PTSD. It is clear that a number of factors, including genetics, influence whether an individual will develop PTSD subsequent to a trauma. The 129S1/SvImJ (S1) inbred mouse strain displays poor fear extinction and may be useful to model this specific aspect of PTSD. The metabotropic glutamate receptor 7 (mGlu7 receptor) has previously been shown to be involved in cognitive processes and anxiety-like behaviour placing it in a key position to regulate fear extinction processes. We sought to compare mGlu7 receptor mRNA levels in the S1 strain with those in the robustly extinguishing C57BL/6J (B6) inbred strain using in situ hybridisation (ISH) in three brain regions associated with fear extinction: the amygdala, hippocampus and prefrontal cortex (PFC). RESULTS Compared to the B6 strain, S1 mice had increased mGlu7 receptor mRNA levels in the lateral amygdala (LA) and basolateral amygdala (BLA) subdivisions. An increase was also seen in the hippocampal CA1 and CA3 subregions of S1 mice. No difference in mGlu7 receptor levels were seen in the central nucleus (CeA) of the amygdala, dentate gyrus (DG) of the hippocampus or prefrontal cortex. CONCLUSIONS These data show altered mGlu7 receptor expression in key brain regions associated with fear extinction in two different inbred mouse strains which differ markedly in their fear extinction behaviour. Altered mGlu7 receptor levels may contribute to the deficit fear extinction processes seen in fear extinction in the S1 strain.
Collapse
Affiliation(s)
- Richard M. O’Connor
- Department of Anatomy and Neuroscience and APC Microbiome Institute, University College Cork, Cork, Ireland,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, S10-20 Hess CSM, 1470 Madison Avenue, New York, NY 10029, USA,Present address: Department of Neuroscience, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Cian P. McCafferty
- Department of Anatomy and Neuroscience and APC Microbiome Institute, University College Cork, Cork, Ireland,Present address: Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Javier A. Bravo
- Grupo de NeuroGastroBioquímica, Laboratorio e Química Biológica & Bioquímica de Sistemas, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA
| | - John F. Cryan
- Department of Anatomy and Neuroscience and APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
23
|
Bossi S, Helleringer R, Galante M, Monlleó E, Trapero A, Rovira X, Daniel H, Llebaria A, McLean H. A Light-Controlled Allosteric Modulator Unveils a Role for mGlu 4 Receptors During Early Stages of Ischemia in the Rodent Cerebellar Cortex. Front Cell Neurosci 2018; 12:449. [PMID: 30542267 PMCID: PMC6277836 DOI: 10.3389/fncel.2018.00449] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/07/2018] [Indexed: 01/15/2023] Open
Abstract
Metabotropic glutamate receptors (mGlus) are G Protein coupled-receptors that modulate synaptic transmission and plasticity in the central nervous system. Some act as autoreceptors to control neurotransmitter release at excitatory synapses and have become attractive targets for drug therapy to treat certain neurological disorders. However, the high degree of sequence conservation around the glutamate binding site makes the development of subtype-specific orthosteric ligands difficult to achieve. This problem can be circumvented by designing molecules that target specific less well conserved allosteric sites. One such allosteric drug, the photo-switchable compound OptoGluNAM4.1, has been recently employed to reversibly inhibit the activity of metabotropic glutamate 4 (mGlu4) receptors in cell cultures and in vivo. We studied OptoGluNAM4.1 as a negative modulator of neurotransmission in rodent cerebellar slices at the parallel fiber – Purkinje cell synapse. Our data show that OptoGluNAM4.1 antagonizes pharmacological activation of mGlu4 receptors in a fully reversible and photo-controllable manner. In addition, for the first time, this new allosteric modulator allowed us to demonstrate that, in brain slices from the rodent cerebellar cortex, mGlu4 receptors are endogenously activated in excitotoxic conditions, such as the early phases of simulated cerebellar ischemia, which is associated with elevated levels of extracellular glutamate. These findings support OptoGluNAM4.1 as a promising new tool for unraveling the role of mGlu4 receptors in the central nervous system in physio-pathological conditions.
Collapse
Affiliation(s)
- Simon Bossi
- Pharmacologie et Biochimie de la Synapse, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud - CNRS, UMR 9197, Orsay, France
| | - Romain Helleringer
- Pharmacologie et Biochimie de la Synapse, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud - CNRS, UMR 9197, Orsay, France
| | - Micaela Galante
- Pharmacologie et Biochimie de la Synapse, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud - CNRS, UMR 9197, Orsay, France
| | - Ester Monlleó
- MCS, Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Ana Trapero
- MCS, Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Xavier Rovira
- Molecular Photopharmacology Research Group, The Tissue Repair and Regeneration Laboratory, University of Vic - Central University of Catalonia, Vic, Spain
| | - Hervé Daniel
- Pharmacologie et Biochimie de la Synapse, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud - CNRS, UMR 9197, Orsay, France
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Heather McLean
- Pharmacologie et Biochimie de la Synapse, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud - CNRS, UMR 9197, Orsay, France
| |
Collapse
|
24
|
Fisher NM, Seto M, Lindsley CW, Niswender CM. Metabotropic Glutamate Receptor 7: A New Therapeutic Target in Neurodevelopmental Disorders. Front Mol Neurosci 2018; 11:387. [PMID: 30405350 PMCID: PMC6206046 DOI: 10.3389/fnmol.2018.00387] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/01/2018] [Indexed: 12/27/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are characterized by a wide range of symptoms including delayed speech, intellectual disability, motor dysfunction, social deficits, breathing problems, structural abnormalities, and epilepsy. Unfortunately, current treatment strategies are limited and innovative new approaches are sorely needed to address these complex diseases. The metabotropic glutamate receptors are a class of G protein-coupled receptors that act to modulate neurotransmission across many brain structures. They have shown great promise as drug targets for numerous neurological and psychiatric diseases. Moreover, the development of subtype-selective allosteric modulators has allowed detailed studies of each receptor subtype. Here, we focus on the metabotropic glutamate receptor 7 (mGlu7) as a potential therapeutic target for NDDs. mGlu7 is expressed widely throughout the brain in regions that correspond to the symptom domains listed above and has established roles in synaptic physiology and behavior. Single nucleotide polymorphisms and mutations in the GRM7 gene have been associated with idiopathic autism and other NDDs in patients. In rodent models, existing literature suggests that decreased mGlu7 expression and/or function may lead to symptoms that overlap with those of NDDs. Furthermore, potentiation of mGlu7 activity has shown efficacy in a mouse model of Rett syndrome. In this review, we summarize current findings that provide rationale for the continued development of mGlu7 modulators as potential therapeutics.
Collapse
Affiliation(s)
- Nicole M Fisher
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Mabel Seto
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Craig W Lindsley
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.,Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Colleen M Niswender
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
25
|
Fisher NM, Gogliotti RG, Vermudez SAD, Stansley BJ, Conn PJ, Niswender CM. Genetic Reduction or Negative Modulation of mGlu 7 Does Not Impact Anxiety and Fear Learning Phenotypes in a Mouse Model of MECP2 Duplication Syndrome. ACS Chem Neurosci 2018; 9:2210-2217. [PMID: 29227625 DOI: 10.1021/acschemneuro.7b00414] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Rett syndrome and MECP2 Duplication syndrome are neurodevelopmental disorders attributed to loss-of-function mutations in, or duplication of, the gene encoding methyl-CpG-binding protein 2 (MeCP2), respectively. We recently reported decreased expression and function of the metabotropic glutamate receptor 7 (mGlu7) in a mouse model of Rett syndrome. Positive allosteric modulation of mGlu7 activity was sufficient to improve several disease phenotypes including cognition. Here, we tested the hypothesis that mGlu7 expression would be reciprocally regulated in a mouse model of MECP2 Duplication syndrome, such that negative modulation of mGlu7 activity would exert therapeutic benefit. To the contrary, we report that mGlu7 is not functionally increased in mice overexpressing MeCP2 and that neither genetic nor pharmacological reduction of mGlu7 activity impacts phenotypes that are antiparallel to those observed in Rett syndrome model mice. These data expand our understanding of how mGlu7 expression and function is affected by changes in MeCP2 dosage and have important implications for the therapeutic development of mGlu7 modulators.
Collapse
Affiliation(s)
- Nicole M. Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Rocco G. Gogliotti
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Sheryl Anne D. Vermudez
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Branden J. Stansley
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Colleen M. Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
26
|
Joffe ME, Centanni SW, Jaramillo AA, Winder DG, Conn PJ. Metabotropic Glutamate Receptors in Alcohol Use Disorder: Physiology, Plasticity, and Promising Pharmacotherapies. ACS Chem Neurosci 2018; 9:2188-2204. [PMID: 29792024 PMCID: PMC6192262 DOI: 10.1021/acschemneuro.8b00200] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Developing efficacious treatments for alcohol use disorder (AUD) has proven difficult. The insidious nature of the disease necessitates a deep understanding of its underlying biology as well as innovative approaches to ameliorate ethanol-related pathophysiology. Excessive ethanol seeking and relapse are generated by long-term changes to membrane properties, synaptic physiology, and plasticity throughout the limbic system and associated brain structures. Each of these factors can be modulated by metabotropic glutamate (mGlu) receptors, a diverse set of G protein-coupled receptors highly expressed throughout the central nervous system. Here, we discuss how different components of the mGlu receptor family modulate neurotransmission in the limbic system and other brain regions involved in AUD etiology. We then describe how these processes are dysregulated following ethanol exposure and speculate about how mGlu receptor modulation might restore such pathophysiological changes. To that end, we detail the current understanding of the behavioral pharmacology of mGlu receptor-directed drug-like molecules in animal models of AUD. Together, this review highlights the prominent position of the mGlu receptor system in the pathophysiology of AUD and provides encouragement that several classes of mGlu receptor modulators may be translated as viable treatment options.
Collapse
Affiliation(s)
- Max E. Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
| | - Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Anel A. Jaramillo
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Danny G. Winder
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
| |
Collapse
|
27
|
Supraphysiological Levels of Oxygen Exposure During the Neonatal Period Impairs Signaling Pathways Required for Learning and Memory. Sci Rep 2018; 8:9914. [PMID: 29967535 PMCID: PMC6028393 DOI: 10.1038/s41598-018-28220-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/04/2018] [Indexed: 12/31/2022] Open
Abstract
Preterm infants often require prolonged oxygen supplementation and are at high risk of neurodevelopmental impairment. We recently reported that adult mice exposed to neonatal hyperoxia (postnatal day [P] 2 to 14) had spatial navigation memory deficits associated with hippocampal shrinkage. The mechanisms by which early oxidative stress impair neurodevelopment are not known. Our objective was to identify early hyperoxia-induced alterations in hippocampal receptors and signaling pathways necessary for memory formation. We evaluated C57BL/6 mouse pups at P14, exposed to either 85% oxygen or air from P2 to 14. We performed targeted analysis of hippocampal ligand-gated ion channels and proteins necessary for memory formation, and global bioinformatic analysis of differentially expressed hippocampal genes and proteins. Hyperoxia decreased hippocampal mGLU7, TrkB, AKT, ERK2, mTORC1, RPS6, and EIF4E and increased α3, α5, and ɤ2 subunits of GABAA receptor and PTEN proteins, although changes in gene expression were not always concordant. Bioinformatic analysis indicated dysfunction in mitochondria and global protein synthesis and translational processes. In conclusion, supraphysiological oxygen exposure reduced proteins necessary for hippocampus-dependent memory formation and may adversely impact hippocampal mitochondrial function and global protein synthesis. These early hippocampal changes may account for memory deficits seen in preterm survivors following prolonged oxygen supplementation.
Collapse
|
28
|
Gogliotti RG, Senter RK, Fisher NM, Adams J, Zamorano R, Walker AG, Blobaum AL, Engers DW, Hopkins CR, Daniels JS, Jones CK, Lindsley CW, Xiang Z, Conn PJ, Niswender CM. mGlu 7 potentiation rescues cognitive, social, and respiratory phenotypes in a mouse model of Rett syndrome. Sci Transl Med 2018; 9:9/403/eaai7459. [PMID: 28814546 DOI: 10.1126/scitranslmed.aai7459] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 01/25/2017] [Accepted: 04/27/2017] [Indexed: 12/20/2022]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the methyl-CpG binding protein 2 (MECP2) gene. The cognitive impairments seen in mouse models of RTT correlate with deficits in long-term potentiation (LTP) at Schaffer collateral (SC)-CA1 synapses in the hippocampus. Metabotropic glutamate receptor 7 (mGlu7) is the predominant mGlu receptor expressed presynaptically at SC-CA1 synapses in adult mice, and its activation on GABAergic interneurons is necessary for induction of LTP. We demonstrate that pathogenic mutations in MECP2 reduce mGlu7 protein expression in brain tissue from RTT patients and in MECP2-deficient mouse models. In rodents, this reduction impairs mGlu7-mediated control of synaptic transmission. We show that positive allosteric modulation of mGlu7 activity restores LTP and improves contextual fear learning, novel object recognition, and social memory. Furthermore, mGlu7 positive allosteric modulation decreases apneas in Mecp2+/- mice, suggesting that mGlu7 may be a potential therapeutic target for multiple aspects of the RTT phenotype.
Collapse
Affiliation(s)
- Rocco G Gogliotti
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Rebecca K Senter
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Jeffrey Adams
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Rocio Zamorano
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Adam G Walker
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Anna L Blobaum
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Darren W Engers
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Corey R Hopkins
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - J Scott Daniels
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Zixiu Xiang
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA. .,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
29
|
Stansley BJ, Conn PJ. The therapeutic potential of metabotropic glutamate receptor modulation for schizophrenia. Curr Opin Pharmacol 2018; 38:31-36. [PMID: 29486374 PMCID: PMC5949078 DOI: 10.1016/j.coph.2018.02.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 02/10/2018] [Indexed: 12/21/2022]
Abstract
Accumulating evidence suggests that a dysregulation of the glutamatergic system exists in the brains of schizophrenia patients. The metabotropic glutamate (mGlu) receptors are being investigated as novel drug targets for this disease, and have shown promise in both preclinical and clinical studies. Activation of mGlu5 receptors may be efficacious for several symptom domains (positive, negative, and cognitive) and the potential for targeting mGlu5 receptors has been bolstered by recent research on mitigating toxicity profiles associated with mGlu5 activation. Additionally, genetic profiling of schizophrenia patients suggests that genes encoding for mGlu1 and mGlu3 receptors are altered, prompting preclinical studies that have demonstrated potential antipsychotic and cognitive enhancing effects of agents that activate mGlu1 and mGlu3 receptors, respectively. Development of subtype-specific drugs for the mGlu receptors, such as allosteric modulators, could provide a path forward for more efficacious and tolerable therapeutics for 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
|
30
|
Martín R, Ferrero JJ, Collado-Alsina A, Aguado C, Luján R, Torres M, Sánchez-Prieto J. Bidirectional modulation of glutamatergic synaptic transmission by metabotropic glutamate type 7 receptors at Schaffer collateral-CA1 hippocampal synapses. J Physiol 2018; 596:921-940. [PMID: 29280494 DOI: 10.1113/jp275371] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/21/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Neurotransmitter release is inhibited by metabotropic glutamate type 7 (mGlu7 ) receptors that reduce Ca2+ influx, yet synapses lacking this receptor also produce weaker release, suggesting that mGlu7 receptors may also prime synaptic vesicles for release. Prolonged activation of mGlu7 receptors with the agonist l-AP4 first reduces and then enhances the amplitude of EPSCs through a presynaptic effect. The inhibitory response is blocked by pertussis toxin, while the potentiating response is prevented by a phospholipase C inhibitor (U73122) and an inhibitor of diacylglycerol (DAG) binding (calphostin C), suggesting that this receptor also couples to pathways that generate DAG. Release potentiation is associated with an increase in the number of synaptic vesicles close to the plasma membrane, which was dependent on the Munc13-2 and RIM1α proteins. The Glu7 receptors activated by the glutamate released following high frequency stimulation provoke a bidirectional modulation of synaptic transmission. ABSTRACT Neurotransmitter release is driven by Ca2+ influx at synaptic boutons that acts on synaptic vesicles ready to undergo exocytosis. Neurotransmitter release is inhibited when metabotropic glutamate type 7 (mGlu7 ) receptors provoke a reduction in Ca2+ influx, although the reduced release from synapses lacking this receptor suggests that they may also prime synaptic vesicles for release. These mGlu7 receptors activate phospholipase C (PLC) and generate inositol trisphosphate, which in turn releases Ca2+ from intracellular stores and produces diacylglycerol (DAG), an activator of proteins containing DAG-binding domains such as Munc13 and protein kinase C (PKC). However, the full effects of mGlu7 receptor signalling on synaptic transmission are unclear. We found that prolonged activation of mGlu7 receptors with the agonist l-AP4 first reduces and then enhances the amplitude of EPSCs, a presynaptic effect that changes the frequency but not the amplitude of the mEPSCs and the paired pulse ratio. Pertussis toxin blocks the inhibitory response, while the PLC inhibitor U73122, and the inhibitor of DAG binding calphostin C, prevent receptor mediated potentiation. Moreover, this DAG-dependent potentiation of the release machinery brings more synaptic vesicles closer to the active zone plasma membrane in a Munc13-2- and RIM1α-dependent manner. Electrically evoked release of glutamate that activates mGlu7 receptors also bidirectionally modulates synaptic transmission. In these conditions, potentiation now occurs rapidly and it overcomes any inhibition, such that potentiation prevails unless it is suppressed with the PLC inhibitor U73122.
Collapse
Affiliation(s)
- Ricardo Martín
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, 28040, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - José Javier Ferrero
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, 28040, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Andrea Collado-Alsina
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, 28040, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Carolina Aguado
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Magdalena Torres
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, 28040, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - José Sánchez-Prieto
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, 28040, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
| |
Collapse
|
31
|
Tresadern G, Trabanco AA, Pérez-Benito L, Overington JP, van Vlijmen HWT, van Westen GJP. Identification of Allosteric Modulators of Metabotropic Glutamate 7 Receptor Using Proteochemometric Modeling. J Chem Inf Model 2017; 57:2976-2985. [PMID: 29172488 PMCID: PMC5755953 DOI: 10.1021/acs.jcim.7b00338] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Indexed: 01/07/2023]
Abstract
Proteochemometric modeling (PCM) is a computational approach that can be considered an extension of quantitative structure-activity relationship (QSAR) modeling, where a single model incorporates information for a family of targets and all the associated ligands instead of modeling activity versus one target. This is especially useful for situations where bioactivity data exists for similar proteins but is scarce for the protein of interest. Here we demonstrate the application of PCM to identify allosteric modulators of metabotropic glutamate (mGlu) receptors. Given our long-running interest in modulating mGlu receptor function we compiled a matrix of compound-target bioactivity data. Some members of the mGlu family are well explored both internally and in the public domain, while there are much fewer examples of ligands for other targets such as the mGlu7 receptor. Using a PCM approach mGlu7 receptor hits were found. In comparison to conventional single target modeling the identified hits were more diverse, had a better confirmation rate, and provide starting points for further exploration. We conclude that the robust structure-activity relationship from well explored target family members translated to better quality hits for PCM compared to virtual screening (VS) based on a single target.
Collapse
Affiliation(s)
- Gary Tresadern
- Computational
Chemistry and Neuroscience Medicinal Chemistry, Janssen
Research & Development, Janssen-Cilag
S.A., Jarama 75A, 45007 Toledo, Spain
| | - Andres A. Trabanco
- Computational
Chemistry and Neuroscience Medicinal Chemistry, Janssen
Research & Development, Janssen-Cilag
S.A., Jarama 75A, 45007 Toledo, Spain
| | - Laura Pérez-Benito
- Computational
Chemistry and Neuroscience Medicinal Chemistry, Janssen
Research & Development, Janssen-Cilag
S.A., Jarama 75A, 45007 Toledo, Spain
| | - John P. Overington
- ChEMBL Group, EMBL-EBI,
Wellcome Trust Genome Campus, CB10 1SD Hinxton, United Kingdom
| | - Herman W. T. van Vlijmen
- Computational
Chemistry, Janssen Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | | |
Collapse
|
32
|
Maksymetz J, Moran SP, Conn PJ. Targeting metabotropic glutamate receptors for novel treatments of schizophrenia. Mol Brain 2017; 10:15. [PMID: 28446243 PMCID: PMC5405554 DOI: 10.1186/s13041-017-0293-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/04/2017] [Indexed: 12/22/2022] Open
Abstract
Support for the N-methyl-D-aspartate receptor (NMDAR) hypofunction hypothesis of schizophrenia has led to increasing focus on restoring proper glutamatergic signaling as an approach for treatment of this devastating disease. The ability of metabotropic glutamate (mGlu) receptors to modulate glutamatergic neurotransmission has thus attracted considerable attention for the development of novel antipsychotics. Consisting of eight subtypes classified into three groups based on sequence homology, signal transduction, and pharmacology, the mGlu receptors provide a wide range of targets to modulate NMDAR function as well as glutamate release. Recently, allosteric modulators of mGlu receptors have been developed that allow unprecedented selectivity among subtypes, not just groups, facilitating the investigation of the effects of subtype-specific modulation. In preclinical animal models, positive allosteric modulators (PAMs) of the group I mGlu receptor mGlu5 have efficacy across all three symptom domains of schizophrenia (positive, negative, and cognitive). The discovery and development of mGlu5 PAMs that display unique signal bias suggests that efficacy can be retained while avoiding the neurotoxic effects of earlier compounds. Interestingly, mGlu1 negative allosteric modulators (NAMs) appear efficacious in positive symptom models of the disease but are still in early preclinical development. While selective group II mGlu receptor (mGlu2/3) agonists have reached clinical trials but were unsuccessful, specific mGlu2 or mGlu3 receptor targeting still hold great promise. Genetic studies implicated mGlu2 in the antipsychotic effects of group II agonists and mGlu2 PAMs have since entered into clinical trials. Additionally, mGlu3 appears to play an important role in cognition, may confer neuroprotective effects, and thus is a promising target to alleviate cognitive deficits in schizophrenia. Although group III mGlu receptors (mGlu4/6/7/8) have attracted less attention, mGlu4 agonists and PAMs appear to have efficacy across all three symptoms domains in preclinical models. The recent discovery of heterodimers comprising mGlu2 and mGlu4 may explain the efficacy of mGlu4 selective compounds but this remains to be determined. Taken together, compounds targeting mGlu receptors, specifically subtype-selective allosteric modulators, provide a compelling alternative approach to fill the unmet clinical needs for patients with schizophrenia.
Collapse
Affiliation(s)
- James Maksymetz
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
- Vanderbilt Center for Neuroscience Drug Discovery, Nashville, TN 37232 USA
| | - Sean P. Moran
- Vanderbilt Center for Neuroscience Drug Discovery, Nashville, TN 37232 USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232 USA
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
- Vanderbilt Center for Neuroscience Drug Discovery, Nashville, TN 37232 USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232 USA
| |
Collapse
|
33
|
Hellyer S, Leach K, Gregory KJ. Neurobiological insights and novel therapeutic opportunities for CNS disorders from mGlu receptor allosteric and biased modulation. Curr Opin Pharmacol 2017; 32:49-55. [DOI: 10.1016/j.coph.2016.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 11/30/2022]
|
34
|
Lindsley CW, Emmitte KA, Hopkins CR, Bridges TM, Gregory KJ, Niswender CM, Conn PJ. Practical Strategies and Concepts in GPCR Allosteric Modulator Discovery: Recent Advances with Metabotropic Glutamate Receptors. Chem Rev 2016; 116:6707-41. [PMID: 26882314 PMCID: PMC4988345 DOI: 10.1021/acs.chemrev.5b00656] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allosteric modulation of GPCRs has initiated a new era of basic and translational discovery, filled with therapeutic promise yet fraught with caveats. Allosteric ligands stabilize unique conformations of the GPCR that afford fundamentally new receptors, capable of novel pharmacology, unprecedented subtype selectivity, and unique signal bias. This review provides a comprehensive overview of the basics of GPCR allosteric pharmacology, medicinal chemistry, drug metabolism, and validated approaches to address each of the major challenges and caveats. Then, the review narrows focus to highlight recent advances in the discovery of allosteric ligands for metabotropic glutamate receptor subtypes 1-5 and 7 (mGlu1-5,7) highlighting key concepts ("molecular switches", signal bias, heterodimers) and practical solutions to enable the development of tool compounds and clinical candidates. The review closes with a section on late-breaking new advances with allosteric ligands for other GPCRs and emerging data for endogenous allosteric modulators.
Collapse
Affiliation(s)
- Craig W. Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kyle A. Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, Texas 76107, United States
| | - Corey R. Hopkins
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville VIC 3052, Australia
| | - Colleen M. Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
35
|
O'Brien DE, Conn PJ. Neurobiological Insights from mGlu Receptor Allosteric Modulation. Int J Neuropsychopharmacol 2016; 19:pyv133. [PMID: 26647381 PMCID: PMC4886670 DOI: 10.1093/ijnp/pyv133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/15/2015] [Accepted: 12/02/2015] [Indexed: 12/25/2022] Open
Abstract
Allosteric modulation of metabotropic glutamate (mGlu) receptors offers a promising pharmacological approach to normalize neural circuit dysfunction associated with various psychiatric and neurological disorders. As mGlu receptor allosteric modulators progress through discovery and clinical development, both technical advances and novel tool compounds are providing opportunities to better understand mGlu receptor pharmacology and neurobiology. Recent advances in structural biology are elucidating the structural determinants of mGlu receptor-negative allosteric modulation and supplying the means to resolve active, allosteric modulator-bound mGlu receptors. The discovery and characterization of allosteric modulators with novel pharmacological profiles is uncovering the biological significance of their intrinsic agonist activity, biased mGlu receptor modulation, and novel mGlu receptor heterodimers. The development and exploitation of optogenetic and optopharmacological tools is permitting a refined spatial and temporal understanding of both mGlu receptor functions and their allosteric modulation in intact brain circuits. Together, these lines of research promise to provide a more refined understanding of mGlu receptors and their allosteric modulation that will inform the development of mGlu receptor allosteric modulators as neurotherapeutics in the years to come.
Collapse
Affiliation(s)
- Daniel E O'Brien
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee (Drs O'Brien and Conn)
| | - P Jeffrey Conn
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee (Drs O'Brien and Conn).
| |
Collapse
|
36
|
Tassin V, Girard B, Chotte A, Fontanaud P, Rigault D, Kalinichev M, Perroy J, Acher F, Fagni L, Bertaso F. Phasic and Tonic mGlu7 Receptor Activity Modulates the Thalamocortical Network. Front Neural Circuits 2016; 10:31. [PMID: 27199672 PMCID: PMC4842779 DOI: 10.3389/fncir.2016.00031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/05/2016] [Indexed: 11/13/2022] Open
Abstract
Mutation of the metabotropic glutamate receptor type 7 (mGlu7) induces absence-like epileptic seizures, but its precise role in the somatosensory thalamocortical network remains unknown. By combining electrophysiological recordings, optogenetics, and pharmacology, we dissected the contribution of the mGlu7 receptor at mouse thalamic synapses. We found that mGlu7 is functionally expressed at both glutamatergic and GABAergic synapses, where it can inhibit neurotransmission and regulate short-term plasticity. These effects depend on the PDZ-ligand of the receptor, as they are lost in mutant mice. Interestingly, the very low affinity of mGlu7 receptors for glutamate raises the question of how it can be activated, namely at GABAergic synapses and in basal conditions. Inactivation of the receptor activity with the mGlu7 negative allosteric modulator (NAM), ADX71743, enhances thalamic synaptic transmission. In vivo administration of the NAM induces a lethargic state with spindle and/or spike-and-wave discharges accompanied by a behavioral arrest typical of absence epileptic seizures. This provides evidence for mGlu7 receptor-mediated tonic modulation of a physiological function in vivo preventing synchronous and potentially pathological oscillations.
Collapse
Affiliation(s)
- Valériane Tassin
- CNRS, Institut de Génomique Fonctionnelle, UMR-5203Montpellier, France; INSERM, U1191Montpellier, France; UMR-5203, Université de MontpellierMontpellier, France
| | - Benoît Girard
- CNRS, Institut de Génomique Fonctionnelle, UMR-5203Montpellier, France; INSERM, U1191Montpellier, France; UMR-5203, Université de MontpellierMontpellier, France
| | - Apolline Chotte
- CNRS, Institut de Génomique Fonctionnelle, UMR-5203Montpellier, France; INSERM, U1191Montpellier, France; UMR-5203, Université de MontpellierMontpellier, France
| | - Pierre Fontanaud
- CNRS, Institut de Génomique Fonctionnelle, UMR-5203Montpellier, France; INSERM, U1191Montpellier, France; UMR-5203, Université de MontpellierMontpellier, France
| | | | | | - Julie Perroy
- CNRS, Institut de Génomique Fonctionnelle, UMR-5203Montpellier, France; INSERM, U1191Montpellier, France; UMR-5203, Université de MontpellierMontpellier, France
| | - Francine Acher
- CNRS, UMR-8601, Université Paris Descartes Paris, France
| | - Laurent Fagni
- CNRS, Institut de Génomique Fonctionnelle, UMR-5203Montpellier, France; INSERM, U1191Montpellier, France; UMR-5203, Université de MontpellierMontpellier, France
| | - Federica Bertaso
- CNRS, Institut de Génomique Fonctionnelle, UMR-5203Montpellier, France; INSERM, U1191Montpellier, France; UMR-5203, Université de MontpellierMontpellier, France
| |
Collapse
|