1
|
Lee SH, Mak A, Verheijen MHG. Comparative assessment of the effects of DREADDs and endogenously expressed GPCRs in hippocampal astrocytes on synaptic activity and memory. Front Cell Neurosci 2023; 17:1159756. [PMID: 37051110 PMCID: PMC10083367 DOI: 10.3389/fncel.2023.1159756] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/06/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) have proven themselves as one of the key in vivo techniques of modern neuroscience, allowing for unprecedented access to cellular manipulations in living animals. With respect to astrocyte research, DREADDs have become a popular method to examine the functional aspects of astrocyte activity, particularly G-protein coupled receptor (GPCR)-mediated intracellular calcium (Ca2+) and cyclic adenosine monophosphate (cAMP) dynamics. With this method it has become possible to directly link the physiological aspects of astrocytic function to cognitive processes such as memory. As a result, a multitude of studies have explored the impact of DREADD activation in astrocytes on synaptic activity and memory. However, the emergence of varying results prompts us to reconsider the degree to which DREADDs expressed in astrocytes accurately mimic endogenous GPCR activity. Here we compare the major downstream signaling mechanisms, synaptic, and behavioral effects of stimulating Gq-, Gs-, and Gi-DREADDs in hippocampal astrocytes of adult mice to those of endogenously expressed GPCRs.
Collapse
Affiliation(s)
- Sophie H. Lee
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Research Master’s Programme Brain and Cognitive Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Aline Mak
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mark H. G. Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: Mark Verheijen,
| |
Collapse
|
2
|
Single cell molecular alterations reveal target cells and pathways of conditioned fear memory. Brain Res 2023; 1807:148309. [PMID: 36870465 DOI: 10.1016/j.brainres.2023.148309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/05/2022] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
OBJECTIVES Recent evidence indicates that hippocampus is important for conditioned fear memory (CFM). Though few studies consider the roles of various cell types' contribution to such a process, as well as the accompanying transcriptome changes during this process. The purpose of this study was to explore the transcriptional regulatory genes and the targeted cells that are altered by CFM reconsolidation. METHODS A fear conditioning experiment was established on adult male C57 mice, after day 3 tone-cued CFM reconsolidation test, hippocampus cells were dissociated. Using single cell RNA sequencing (scRNA-seq) technique, alterations of transcriptional genes expression were detected and cell cluster analysis were performed and compared with those in sham group. RESULTS Seven non-neuronal and eight neuronal cell clusters (including four known neurons and four newly identified neuronal subtypes) has been explored. Among them, CA subtype 1 has characteristic gene markers of Ttr and Ptgds, which is speculated to be the outcome of acute stress and promotes the production of CFM. The results of KEGG pathway enrichment indicate the differences in the expression of certain molecular protein functional subunits in long-term potentiation (LTP) pathway between two types of neurons (DG and CA1) and astrocytes, thus providing a new transcriptional perspective for the role of hippocampus in the CFM reconsolidation. More importantly, the correlation between the reconsolidation of CFM and neurodegenerative diseases-linked genes is substantiated by the results from cell-cell interactions and KEGG pathway enrichment. Further analysis shows that the reconsolidation of CFM inhibits the risk-factor genes App and ApoE in Alzheimer's Disease (AD) and activates the protective gene Lrp1. CONCLUSIONS This study reports the transcriptional genes expression changes of hippocampal cells driven by CFM, which confirm the involvement of LTP pathway and suggest the possibility of CFM-like behavior in preventing AD. However, the current research is limited to normal C57 mice, and further studies on AD model mice are needed to prove this preliminary conclusion.
Collapse
|
3
|
Dogra S, Putnam J, Conn PJ. Metabotropic glutamate receptor 3 as a potential therapeutic target for psychiatric and neurological disorders. Pharmacol Biochem Behav 2022; 221:173493. [PMID: 36402243 PMCID: PMC9729465 DOI: 10.1016/j.pbb.2022.173493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/22/2022] [Revised: 11/08/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022]
Abstract
Glutamate is a major excitatory neurotransmitter in the central nervous system (CNS) and abnormalities in the glutamatergic system underlie various CNS disorders. As metabotropic glutamate receptor 3 (mGlu3 receptor) regulates glutamatergic transmission in various brain areas, emerging literature suggests that targeting mGlu3 receptors can be a novel approach to the treatment of psychiatric and neurological disorders. For example, mGlu3 receptor negative allosteric modulators (NAMs) induce rapid antidepressant-like effects in both acute and chronic stress models. Activation of mGlu3 receptors can enhance cognition in the rodents modeling schizophrenia-like pathophysiology. The mGlu3 receptors expressed in the astrocytes induce neuroprotective effects. Although polymorphisms in GRM3 have been shown to be associated with addiction, there is not significant evidence about the efficacy of mGlu3 receptor ligands in rodent models of addiction. Collectively, drugs targeting mGlu3 receptors may provide an alternative approach to fill the unmet clinical need for safer and more efficacious therapeutics for CNS disorders.
Collapse
Affiliation(s)
- Shalini Dogra
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Jason Putnam
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
4
|
Tyler RE, Besheer J, Joffe ME. Advances in translating mGlu 2 and mGlu 3 receptor selective allosteric modulators as breakthrough treatments for affective disorders and alcohol use disorder. Pharmacol Biochem Behav 2022; 219:173450. [PMID: 35988792 PMCID: PMC10405528 DOI: 10.1016/j.pbb.2022.173450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/01/2022] [Revised: 07/26/2022] [Accepted: 08/13/2022] [Indexed: 11/16/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are promising targets for the treatment of affective disorders and alcohol use disorder (AUD). Nonspecific ligands for Group II (mGlu2 and mGlu3) mGlu receptors have demonstrated consistent therapeutic potential for affective disorders in preclinical models. Disentangling the specific roles of mGlu2 versus mGlu3 receptors in these effects has persisted as a major challenge, in part due to pharmacological limitations. However, the recent development of highly specific allosteric modulators for both mGlu2 and mGlu3 receptors have enabled straightforward and rigorous investigations into the specific function of each receptor. Here, we review recent experiments using these compounds that have demonstrated both similar and distinct receptor functions in behavioral, molecular, and electrophysiological measures associated with basal function and preclinical models of affective disorders. Studies using these selective drugs have demonstrated that mGlu2 is the predominant receptor subclass involved in presynaptic neurotransmitter release in prefrontal cortex. By contrast, the activation of postsynaptic mGlu3 receptors induces a cascade of cellular changes that results in AMPA receptor internalization, producing long-term depression and diminishing excitatory drive. Acute stress decreases the mGlu3 receptor function and dynamically alters transcript expression for both mGlu2 (Grm2) and mGlu3 (Grm3) receptors in brain areas involved in reward and stress. Accordingly, both mGlu2 and mGlu3 negative allosteric modulators show acute antidepressant-like effects and potential prophylactic effects against acute and traumatic stressors. The wide array of effects displayed by these new allosteric modulators of mGlu2 and mGlu3 receptors suggest that these drugs may act through improving endophenotypes of symptoms observed across several neuropsychiatric disorders. Therefore, recently developed allosteric modulators selective for mGlu2 or mGlu3 receptors show promise as potential therapeutics for affective disorders and AUD.
Collapse
Affiliation(s)
- Ryan E Tyler
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Neuroscience Curriculum, University of North Carolina at Chapel Hill, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, USA
| | - Joyce Besheer
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Neuroscience Curriculum, University of North Carolina at Chapel Hill, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, USA
| | - Max E Joffe
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219, USA; Translational Neuroscience Program, University of Pittsburgh, USA.
| |
Collapse
|
5
|
Exploration of group II metabotropic glutamate receptor modulation in mouse models of Rett syndrome and MECP2 Duplication syndrome. Neuropharmacology 2022; 209:109022. [PMID: 35248529 PMCID: PMC8973998 DOI: 10.1016/j.neuropharm.2022.109022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/20/2021] [Revised: 02/11/2022] [Accepted: 02/27/2022] [Indexed: 01/01/2023]
Abstract
Rett syndrome (RTT) and MECP2 Duplication syndrome (MDS) have opposing molecular origins in relation to expression and function of the transcriptional regulator Methyl-CpG-binding protein 2 (MeCP2). Several clinical and preclinical phenotypes, however, are shared between these disorders. Modulation of MeCP2 levels has recently emerged as a potential treatment option for both of these diseases. However, toxicity concerns remain with these approaches. Here, we focus on pharmacologically modulating the group II metabotropic glutamate receptors (mGlu), mGlu2 and mGlu3, which are two downstream targets of MeCP2 that are bidirectionally affected in expression in RTT patients and mice (Mecp2Null/+) versus an MDS mouse model (MECP2Tg1/o). Mecp2Null/+ and MECP2Tg1/o animals also exhibit contrasting phenotypes in trace fear acquisition, a form of temporal associative learning and memory, with trace fear deficiency observed in Mecp2Null/+ mice and abnormally enhanced trace fear acquisition in MECP2Tg1/o animals. In Mecp2Null/+ mice, treatment with the mGlu2/3 agonist LY379268 reverses the deficit in trace fear acquisition, and mGlu2/3 antagonism with LY341495 normalizes the abnormal trace fear learning and memory phenotype in MECP2Tg1/o mice. Altogether, these data highlight the role of group II mGlu receptors in RTT and MDS and demonstrate that both mGlu2 and mGlu3 may be potential therapeutic targets for these disorders.
Collapse
|
6
|
Veselinović T, Neuner I. Progress and Pitfalls in Developing Agents to Treat Neurocognitive Deficits Associated with Schizophrenia. CNS Drugs 2022; 36:819-858. [PMID: 35831706 PMCID: PMC9345797 DOI: 10.1007/s40263-022-00935-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Accepted: 06/06/2022] [Indexed: 12/11/2022]
Abstract
Cognitive impairments associated with schizophrenia (CIAS) represent a central element of the symptomatology of this severe mental disorder. CIAS substantially determine the disease prognosis and hardly, if at all, respond to treatment with currently available antipsychotics. Remarkably, all drugs presently approved for the treatment of schizophrenia are, to varying degrees, dopamine D2/D3 receptor blockers. In turn, rapidly growing evidence suggests the immense significance of systems other than the dopaminergic system in the genesis of CIAS. Accordingly, current efforts addressing the unmet needs of patients with schizophrenia are primarily based on interventions in other non-dopaminergic systems. In this review article, we provide a brief overview of the available evidence on the importance of specific systems in the development of CIAS. In addition, we describe the promising targets for the development of new drugs that have been used so far. In doing so, we present the most important candidates that have been investigated in the field of the specific systems in recent years and present a summary of the results available at the time of drafting this review (May 2022), as well as the currently ongoing studies.
Collapse
Affiliation(s)
- Tanja Veselinović
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany.
| | - Irene Neuner
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- JARA-BRAIN, Aachen, Germany
| |
Collapse
|
7
|
Targeting metabotropic glutamate receptors for the treatment of depression and other stress-related disorders. Neuropharmacology 2021; 196:108687. [PMID: 34175327 DOI: 10.1016/j.neuropharm.2021.108687] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/11/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022]
Abstract
The discovery of robust antidepressant effects of ketamine in refractory patients has led to increasing focus on agents targeting glutamatergic signaling as potential novel antidepressant strategy. Among the agents targeting the glutamatergic system, compounds acting at metabotropic glutamate (mGlu) receptors are among the most promising agents under studies for depressive disorders. Further, the receptor diversity, distinct distribution in the CNS, and ability to modulate the glutamatergic neurotransmission in the brain areas implicated in mood disorders make them an exciting target for stress-related disorders. In preclinical models, antidepressant and anxiolytic effects of mGlu5 negative allosteric modulators (NAMs) have been reported. Interestingly, mGlu2/3 receptor antagonists show fast and sustained antidepressant-like effects similar to that of ketamine in rodents. Excitingly, they can also induce antidepressant effects in the animal models of treatment-resistant depression and are devoid of the side-effects associated with ketamine. Unfortunately, clinical trials of both mGlu5 and mGlu2/3 receptor NAMs have been inconclusive, and additional trials using other compounds with suitable preclinical and clinical properties are needed. Although group III mGlu receptors have gained less attention, mGlu7 receptor ligands have been shown to induce antidepressant-like effects in rodents. Collectively, compounds targeting mGlu receptors provide an alternative approach to fill the outstanding clinical need for safer and more efficacious antidepressants. This article is part of the special Issue on "Glutamate Receptors - mGluRs".
Collapse
|
8
|
Egerton A, Grace AA, Stone J, Bossong MG, Sand M, McGuire P. Glutamate in schizophrenia: Neurodevelopmental perspectives and drug development. Schizophr Res 2020; 223:59-70. [PMID: 33071070 DOI: 10.1016/j.schres.2020.09.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/19/2019] [Revised: 08/12/2020] [Accepted: 09/20/2020] [Indexed: 12/14/2022]
Abstract
Research into the neurobiological processes that may lead to the onset of schizophrenia places growing emphasis on the glutamatergic system and brain development. Preclinical studies have shown that neurodevelopmental, genetic, and environmental factors contribute to glutamatergic dysfunction and schizophrenia-related phenotypes. Clinical research has suggested that altered brain glutamate levels may be present before the onset of psychosis and relate to outcome in those at clinical high risk. After psychosis onset, glutamate dysfunction may also relate to the degree of antipsychotic response and clinical outcome. These findings support ongoing efforts to develop pharmacological interventions that target the glutamate system and could suggest that glutamatergic compounds may be more effective in specific patient subgroups or illness stages. In this review, we consider the updated glutamate hypothesis of schizophrenia, from a neurodevelopmental perspective, by reviewing recent preclinical and clinical evidence, and discuss the potential implications for novel therapeutics.
Collapse
Affiliation(s)
- Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Stone
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Matthijs G Bossong
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michael Sand
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
9
|
Hasselmo ME, Alexander AS, Hoyland A, Robinson JC, Bezaire MJ, Chapman GW, Saudargiene A, Carstensen LC, Dannenberg H. The Unexplored Territory of Neural Models: Potential Guides for Exploring the Function of Metabotropic Neuromodulation. Neuroscience 2020; 456:143-158. [PMID: 32278058 DOI: 10.1016/j.neuroscience.2020.03.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/10/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/16/2022]
Abstract
The space of possible neural models is enormous and under-explored. Single cell computational neuroscience models account for a range of dynamical properties of membrane potential, but typically do not address network function. In contrast, most models focused on network function address the dimensions of excitatory weight matrices and firing thresholds without addressing the complexities of metabotropic receptor effects on intrinsic properties. There are many under-explored dimensions of neural parameter space, and the field needs a framework for representing what has been explored and what has not. Possible frameworks include maps of parameter spaces, or efforts to categorize the fundamental elements and molecules of neural circuit function. Here we review dimensions that are under-explored in network models that include the metabotropic modulation of synaptic plasticity and presynaptic inhibition, spike frequency adaptation due to calcium-dependent potassium currents, and afterdepolarization due to calcium-sensitive non-specific cation currents and hyperpolarization activated cation currents. Neuroscience research should more effectively explore possible functional models incorporating under-explored dimensions of neural function.
Collapse
Affiliation(s)
- Michael E Hasselmo
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States.
| | - Andrew S Alexander
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Alec Hoyland
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Jennifer C Robinson
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Marianne J Bezaire
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - G William Chapman
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Ausra Saudargiene
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Lucas C Carstensen
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Holger Dannenberg
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| |
Collapse
|
10
|
Heilig M, Augier E, Pfarr S, Sommer WH. Developing neuroscience-based treatments for alcohol addiction: A matter of choice? Transl Psychiatry 2019; 9:255. [PMID: 31594920 PMCID: PMC6783461 DOI: 10.1038/s41398-019-0591-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/15/2019] [Revised: 07/05/2019] [Accepted: 08/08/2019] [Indexed: 12/15/2022] Open
Abstract
Excessive alcohol use is the cause of an ongoing public health crisis, and accounts for ~5% of global disease burden. A minority of people with recreational alcohol use develop alcohol addiction (hereafter equated with "alcohol dependence" or simply "alcoholism"), a condition characterized by a systematically biased choice preference for alcohol at the expense of healthy rewards, and continued use despite adverse consequences ("compulsivity"). Alcoholism is arguably the most pressing area of unmet medical needs in psychiatry, with only a small fraction of patients receiving effective, evidence-based treatments. Medications currently approved for the treatment of alcoholism have small effect sizes, and their clinical uptake is negligible. No mechanistically new medications have been approved since 2004, and promising preclinical results have failed to translate into novel treatments. This has contributed to a reemerging debate whether and to what extent alcohol addiction represents a medical condition, or reflects maladaptive choices without an underlying brain pathology. Here, we review this landscape, and discuss the challenges, lessons learned, and opportunities to retool drug development in this important therapeutic area.
Collapse
Affiliation(s)
- Markus Heilig
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, S-581 83, Linköping, Sweden.
| | - Eric Augier
- 0000 0001 2162 9922grid.5640.7Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, S-581 83 Linköping, Sweden
| | - Simone Pfarr
- 0000 0004 0477 2235grid.413757.3Institute of Psychopharmacology, Central Institute of Mental Health (CIMH), J 5, 68159 Mannheim, Germany
| | - Wolfgang H. Sommer
- 0000 0004 0477 2235grid.413757.3Institute of Psychopharmacology, Central Institute of Mental Health (CIMH), J 5, 68159 Mannheim, Germany ,0000 0004 0477 2235grid.413757.3Department of Addiction Medicine, Central Institute of Mental Health (CIMH), J 5, 68159 Mannheim, Germany
| |
Collapse
|
11
|
Yamada Y, Yohn SE, Gilliland K, Loch MT, Schulte ML, Rodriguez AL, Blobaum AL, Niswender CM, Conn PJ, Lindsley CW. Further exploration of an N-aryl phenoxyethoxy pyridinone-based series of mGlu 3 NAMs: Challenging SAR, enantiospecific activity and in vivo efficacy. Bioorg Med Chem Lett 2019; 29:2670-2674. [PMID: 31358468 DOI: 10.1016/j.bmcl.2019.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/06/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 11/17/2022]
Abstract
This letter describes the further optimization of a series of mGlu3 NAMs based on an N-aryl phenoxyethoxy pyridinone core. A multidimensional optimization campaign, with focused matrix libraries, quickly established challenging SAR, enantiospecific activity, differences in assay read-outs (Ca2+ flux via a promiscuous G protein (Gα15) versus native coupling to GIRK channels), identified both full and partial mGlu3 NAMs and a new in vivo tool compound, VU6017587. This mGlu3 NAM showed efficacy in tail suspension, elevated zero maze and marble burying, suggesting selective inhibition of mGlu3 affords anxiolytic-like and antidepressant-like phenotypes in mice.
Collapse
Affiliation(s)
- Yosuke Yamada
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Samantha E Yohn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Kristen Gilliland
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Mathew T Loch
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Michael L Schulte
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Alice L Rodriguez
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Anna L Blobaum
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
12
|
Stansley BJ, Conn PJ. Neuropharmacological Insight from Allosteric Modulation of mGlu Receptors. Trends Pharmacol Sci 2019; 40:240-252. [PMID: 30824180 PMCID: PMC6445545 DOI: 10.1016/j.tips.2019.02.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/11/2019] [Revised: 02/06/2019] [Accepted: 02/06/2019] [Indexed: 12/11/2022]
Abstract
The metabotropic glutamate (mGlu) receptors are a family of G-protein-coupled receptors (GPCRs) that regulate cell physiology throughout the nervous system. The potential of mGlu receptors as therapeutic targets has been bolstered by current research that has provided insight into the diverse modes of mGlu activation and signaling. In particular, the allosteric modulation of mGlu receptors represents a major area of focus in studies of basic pharmacology as well as drug development, largely due to the high subtype specificity achievable by targeting allosteric sites on mGlu receptors. These provide sophisticated regulation of neuronal excitability and synaptic transmission to influence behavioral output. Here, we review how these allosteric mechanisms have been leveraged preclinically to demonstrate the therapeutic potential of allosteric modulators for neurological and neuropsychiatric disorders, such as autism, cognitive impairment, Parkinson's disease (PD), stress, and schizophrenia.
Collapse
Affiliation(s)
- Branden J Stansley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
13
|
de la Fuente Revenga M, Ibi D, Cuddy T, Toneatti R, Kurita M, Ijaz MK, Miles MF, Wolstenholme JT, González-Maeso J. Chronic clozapine treatment restrains via HDAC2 the performance of mGlu2 receptor agonism in a rodent model of antipsychotic activity. Neuropsychopharmacology 2019; 44:443-454. [PMID: 30038413 PMCID: PMC6300555 DOI: 10.1038/s41386-018-0143-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/22/2018] [Revised: 06/06/2018] [Accepted: 06/25/2018] [Indexed: 01/25/2023]
Abstract
Preclinical findings in rodent models pointed toward activation of metabotropic glutamate 2/3 (mGlu2/3) receptors as a new pharmacological approach to treat psychosis. However, more recent studies failed to show clinical efficacy of mGlu2/3 receptor agonism in schizophrenia patients. We previously proposed that long-term antipsychotic medication restricted the therapeutic effects of these glutamatergic agents. However, little is known about the molecular mechanism underlying the potential repercussion of previous antipsychotic exposure on the therapeutic performance of mGlu2/3 receptor agonists. Here we show that this maladaptive effect of antipsychotic treatment is mediated mostly via histone deacetylase 2 (HDAC2). Chronic treatment with the antipsychotic clozapine led to a decrease in mouse frontal cortex mGlu2 mRNA, an effect that required expression of both HDAC2 and the serotonin 5-HT2A receptor. This transcriptional alteration occurred in association with HDAC2-dependent repressive histone modifications at the mGlu2 promoter. We found that chronic clozapine treatment decreased via HDAC2 the capabilities of the mGlu2/3 receptor agonist LY379268 to activate G-proteins in the frontal cortex of mice. Chronic clozapine treatment blunted the antipsychotic-related behavioral effects of LY379268, an effect that was not observed in HDAC2 knockout mice. More importantly, co-administration of the class I and II HDAC inhibitor SAHA (vorinostat) preserved the antipsychotic profile of LY379268 and frontal cortex mGlu2/3 receptor density in wild-type mice. These findings raise concerns on the design of previous clinical studies with mGlu2/3 agonists, providing the rationale for the development of HDAC2 inhibitors as a new epigenetic-based approach to improve the currently limited response to treatment with glutamatergic antipsychotics.
Collapse
Affiliation(s)
- Mario de la Fuente Revenga
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Daisuke Ibi
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA ,0000 0001 0670 2351grid.59734.3cDepartment Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,grid.259879.8Department of Chemical Pharmacology, Meijo University, Nagoya, 468-8503 Japan
| | - Travis Cuddy
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Rudy Toneatti
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Mitsumasa Kurita
- 0000 0001 0670 2351grid.59734.3cDepartment Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0004 1797 168Xgrid.417741.0Present Address: Dainippon Sumitomo Pharma Co., Ltd., Osaka, 564-0053 Japan
| | - Maryum K. Ijaz
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Michael F. Miles
- 0000 0004 0458 8737grid.224260.0Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA ,0000 0004 0458 8737grid.224260.0VCU Alcohol Research Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Jennifer T. Wolstenholme
- 0000 0004 0458 8737grid.224260.0Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA ,0000 0004 0458 8737grid.224260.0VCU Alcohol Research Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Javier González-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA. .,Department Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
14
|
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] [Academic Contribution 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
|
15
|
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] [Academic Contribution 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
|