1
|
McClatchy DB, Powell SB, Yates JR. In vivo mapping of protein-protein interactions of schizophrenia risk factors generates an interconnected disease network. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571320. [PMID: 38168169 PMCID: PMC10759996 DOI: 10.1101/2023.12.12.571320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Genetic analyses of Schizophrenia (SCZ) patients have identified thousands of risk factors. In silico protein-protein interaction (PPI) network analysis has provided strong evidence that disrupted PPI networks underlie SCZ pathogenesis. In this study, we performed in vivo PPI analysis of several SCZ risk factors in the rodent brain. Using endogenous antibody immunoprecipitations coupled to mass spectrometry (MS) analysis, we constructed a SCZ network comprising 1612 unique PPI with a 5% FDR. Over 90% of the PPI were novel, reflecting the lack of previous PPI MS studies in brain tissue. Our SCZ PPI network was enriched with known SCZ risk factors, which supports the hypothesis that an accumulation of disturbances in selected PPI networks underlies SCZ. We used Stable Isotope Labeling in Mammals (SILAM) to quantitate phencyclidine (PCP) perturbations in the SCZ network and found that PCP weakened most PPI but also led to some enhanced or new PPI. These findings demonstrate that quantitating PPI in perturbed biological states can reveal alterations to network biology.
Collapse
|
2
|
Asch RH, Hillmer AT, Baldassarri SR, Esterlis I. The metabotropic glutamate receptor 5 as a biomarker for psychiatric disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:265-310. [PMID: 36868631 DOI: 10.1016/bs.irn.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of glutamate system in the etiology and pathophysiology of psychiatric disorders has gained considerable attention in the past two decades, including dysregulation of the metabotropic glutamatergic receptor subtype 5 (mGlu5). Thus, mGlu5 may represent a promising therapeutic target for psychiatric conditions, particularly stress-related disorders. Here, we describe mGlu5 findings in mood disorders, anxiety, and trauma disorders, as well as substance use (specifically nicotine, cannabis, and alcohol use). We highlight insights gained from positron emission tomography (PET) studies, where possible, and discuss findings from treatment trials, when available, to explore the role of mGlu5 in these psychiatric disorders. Through the research evidence reviewed in this chapter, we make the argument that, not only is dysregulation of mGlu5 evident in numerous psychiatric disorders, potentially functioning as a disease "biomarker," the normalization of glutamate neurotransmission via changes in mGlu5 expression and/or modulation of mGlu5 signaling may be a needed component in treating some psychiatric disorders or symptoms. Finally, we hope to demonstrate the utility of PET as an important tool for investigating mGlu5 in disease mechanisms and treatment response.
Collapse
Affiliation(s)
- Ruth H Asch
- Department of Psychiatry, Yale University, New Haven, CT, United States.
| | - Ansel T Hillmer
- Department of Psychiatry, Yale University, New Haven, CT, United States; Department of Radiology and Biomedical Imaging, New Haven, CT, United States
| | - Stephen R Baldassarri
- Yale Program in Addiction Medicine, Yale University, New Haven, CT, United States; Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Irina Esterlis
- Department of Psychiatry, Yale University, New Haven, CT, United States; Department of Psychology, Yale University, New Haven, CT, United States; Clinical Neurosciences Division, U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
3
|
Matrisciano F, Locci V, Dong E, Nicoletti F, Guidotti A, Grayson DR. Altered Expression and In Vivo Activity of mGlu5 Variant a Receptors in the Striatum of BTBR Mice: Novel Insights Into the Pathophysiology of Adult Idiopathic Forms of Autism Spectrum Disorders. Curr Neuropharmacol 2022; 20:2354-2368. [PMID: 35139800 PMCID: PMC9890299 DOI: 10.2174/1567202619999220209112609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND mGlu5 metabotropic glutamate receptors are considered as candidate drug targets in the treatment of "monogenic" forms of autism spectrum disorders (ASD), such as Fragile- X syndrome (FXS). However, despite promising preclinical data, clinical trials using mGlu5 receptor antagonists to treat FXS showed no beneficial effects. OBJECTIVE Here, we studied the expression and function of mGlu5 receptors in the striatum of adult BTBR mice, which model idiopathic forms of ASD, and behavioral phenotype. METHODS Behavioral tests were associated with biochemistry analysis including qPCR and western blot for mRNA and protein expression. In vivo analysis of polyphosphoinositides hydrolysis was performed to study the mGlu5-mediated intracellular signaling in the striatum of adult BTBR mice under basal conditions and after MTEP exposure. RESULTS Expression of mGlu5 receptors and mGlu5 receptor-mediated polyphosphoinositides hydrolysis were considerably high in the striatum of BTBR mice, sensitive to MTEP treatment. Changes in the expression of genes encoding for proteins involved in excitatory and inhibitory neurotransmission and synaptic plasticity, including Fmr1, Dlg4, Shank3, Brd4, bdnf-exon IX, Mef2c, and Arc, GriA2, Glun1, Nr2A, and Grm1, Grm2, GriA1, and Gad1 were also found. Behaviorally, BTBR mice showed high repetitive stereotypical behaviors, including self-grooming and deficits in social interactions. Acute or repeated injections with MTEP reversed the stereotyped behavior and the social interaction deficit. Similar effects were observed with the NMDA receptor blockers MK-801 or ketamine. CONCLUSION These findings support a pivotal role of mGlu5 receptor abnormal expression and function in idiopathic ASD adult forms and unveil novel potential targets for therapy.
Collapse
Affiliation(s)
- Francesco Matrisciano
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Valentina Locci
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Erbo Dong
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Center for Alcohol Research in Epigenetics Department of Psychiatry College of Medicine University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Alessandro Guidotti
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Center for Alcohol Research in Epigenetics Department of Psychiatry College of Medicine University of Illinois Chicago, Chicago, IL 60612, USA
| | - Dennis R. Grayson
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Center for Alcohol Research in Epigenetics Department of Psychiatry College of Medicine University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
4
|
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
|
5
|
Yao C, Jiang X, Ye X, Xie T, Bai R. Antidepressant Drug Discovery and Development: Mechanism and Drug Design Based on Small Molecules. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Chuansheng Yao
- School of Pharmacy Hangzhou Normal University Hangzhou 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Xiaoying Jiang
- College of Material, Chemistry and Chemical Engineering Key Laboratory of Organosilicon Chemistry and Material Technology Ministry of Education, Hangzhou Normal University Hangzhou 311121 P.R. China
| | - Xiang‐Yang Ye
- School of Pharmacy Hangzhou Normal University Hangzhou 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Tian Xie
- School of Pharmacy Hangzhou Normal University Hangzhou 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Renren Bai
- School of Pharmacy Hangzhou Normal University Hangzhou 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| |
Collapse
|
6
|
Tsotsokou G, Nikolakopoulou M, Kouvelas ED, Mitsacos A. Neonatal maternal separation affects metabotropic glutamate receptor 5 expression and anxiety-related behavior of adult rats. Eur J Neurosci 2021; 54:4550-4564. [PMID: 34137089 DOI: 10.1111/ejn.15358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 01/15/2023]
Abstract
Exposure to early life stress leads to long-term neurochemical and behavioral alterations. Stress-induced psychiatric disorders, such as depression, have recently been linked to dysregulation of glutamate signaling, mainly via its postsynaptic receptors. The role of metabotropic glutamate receptor 5 (mGluR5) in stress-induced psychopathology has been the target of several studies in humans. In rodents, blockade of mGluR5 produces antidepressant-like actions, whereas mice lacking mGluR5 exhibit altered anxiety-like behaviors and learning. In this study, we used well-known rodent models of early life stress based on mother-infant separation during the first 3 weeks of life in order to examine the effects of neonatal maternal separation on mGluR5 expression and on anxiety-related behavior in adulthood. We observed that brief (15 min) neonatal maternal separation, but not prolonged (3 h), induced increases in mGluR5 mRNA and protein expression levels in medial prefrontal cortex and mGluR5 protein levels in dorsal, but not ventral, hippocampus of adult rat brain. Behavioral testing using the open-field and the elevated-plus maze tasks showed that brief maternal separations resulted in increased exploratory and decreased anxiety-related behavior, whereas prolonged maternal separations resulted in increased anxiety-related behavior in adulthood. The data indicate that the long-lasting effects of neonatal mother-offspring separation on anxiety-like behavior and mGluR5 expression depend on the duration of maternal separation and suggest that the increased mGluR5 receptors in medial prefrontal cortex and hippocampus of adult rats exposed to brief neonatal maternal separations may underlie their heightened ability to cope with stress.
Collapse
Affiliation(s)
- Giota Tsotsokou
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| | - Maria Nikolakopoulou
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| | - Elias D Kouvelas
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| | - Ada Mitsacos
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| |
Collapse
|
7
|
Imbriglio T, Verhaeghe R, Antenucci N, Maccari S, Battaglia G, Nicoletti F, Cannella M. Developmental up-regulation of NMDA receptors in the prefrontal cortex and hippocampus of mGlu5 receptor knock-out mice. Mol Brain 2021; 14:77. [PMID: 33962661 PMCID: PMC8106212 DOI: 10.1186/s13041-021-00784-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/21/2021] [Indexed: 12/02/2022] Open
Abstract
mGlu5 metabotropic glutamate receptors are highly expressed and functional in the early postnatal life, and are known to positively modulate NMDA receptor function. Here, we examined the expression of NMDA receptor subunits and interneuron-related genes in the prefrontal cortex and hippocampus of mGlu5-/- mice and wild-type littermates at three developmental time points (PND9, - 21, and - 75). We were surprised to find that expression of all NMDA receptor subunits was greatly enhanced in mGlu5-/- mice at PND21. In contrast, at PND9, expression of the GluN2B subunit was enhanced, whereas expression of GluN2A and GluN2D subunits was reduced in both regions. These modifications were transient and disappeared in the adult life (PND75). Changes in the transcripts of interneuron-related genes (encoding parvalbumin, somatostatin, vasoactive intestinal peptide, reelin, and the two isoforms of glutamate decarboxylase) were also observed in mGlu5-/- mice across postnatal development. For example, the transcript encoding parvalbumin was up-regulated in the prefrontal cortex of mGlu5-/- mice at PND9 and PND21, whereas it was significantly reduced at PND75. These findings suggest that in mGlu5-/- mice a transient overexpression of NMDA receptor subunits may compensate for the lack of the NMDA receptor partner, mGlu5. Interestingly, in mGlu5-/- mice the behavioral response to the NMDA channel blocker, MK-801, was significantly increased at PND21, and largely reduced at PND75. The impact of adaptive changes in the expression of NMDA receptor subunits should be taken into account when mGlu5-/- mice are used for developmental studies.
Collapse
Affiliation(s)
| | | | - Nico Antenucci
- Department of Physiology and Pharmacology "V. Erspamer", University Sapienza of Rome, Piazzale Aldo Moro, 5, 00185, Rome, Italy
| | - Stefania Maccari
- Department of Physiology and Pharmacology "V. Erspamer", University Sapienza of Rome, Piazzale Aldo Moro, 5, 00185, Rome, Italy
- CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, University of Lille, Lille, France
| | - Giuseppe Battaglia
- IRCCS Neuromed, Pozzilli, IS, Italy
- Department of Physiology and Pharmacology "V. Erspamer", University Sapienza of Rome, Piazzale Aldo Moro, 5, 00185, Rome, Italy
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, IS, Italy.
- Department of Physiology and Pharmacology "V. Erspamer", University Sapienza of Rome, Piazzale Aldo Moro, 5, 00185, Rome, Italy.
| | | |
Collapse
|
8
|
Li W, Kutas M, Gray JA, Hagerman RH, Olichney JM. The Role of Glutamate in Language and Language Disorders - Evidence from ERP and Pharmacologic Studies. Neurosci Biobehav Rev 2020; 119:217-241. [PMID: 33039453 PMCID: PMC11584167 DOI: 10.1016/j.neubiorev.2020.09.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 08/10/2020] [Accepted: 09/21/2020] [Indexed: 12/31/2022]
Abstract
Current models of language processing do not address mechanisms at the neurotransmitter level, nor how pharmacologic agents may improve language function(s) in seemingly disparate disorders. L-Glutamate, the primary excitatory neurotransmitter in the human brain, is extensively involved in various higher cortical functions. We postulate that the physiologic role of L-Glutamate neurotransmission extends to the regulation of language access, comprehension, and production, and that disorders in glutamatergic transmission and circuitry contribute to the pathogenesis of neurodegenerative diseases and sporadic-onset language disorders such as the aphasic stroke syndromes. We start with a review of basic science data pertaining to various glutamate receptors in the CNS and ways that they may influence the physiological processes of language access and comprehension. We then focus on the dysregulation of glutamate neurotransmission in three conditions in which language dysfunction is prominent: Alzheimer's Disease, Fragile X-associated Tremor/Ataxia Syndrome, and Aphasic Stroke Syndromes. Finally, we review the pharmacologic and electrophysiologic (event related brain potential or ERP) data pertaining to the role glutamate neurotransmission plays in language processing and disorders.
Collapse
Affiliation(s)
- Wentao Li
- Department of Neurology, University of California, Davis, 4860 Y Street, Suite 3700, Sacramento, CA, 95817, USA.
| | - Marta Kutas
- Department of Cognitive Science, University of California, San Diego, 9500 Gilman Drive #0515, La Jolla, CA, 92093, USA; Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - John A Gray
- Department of Neurology, University of California, Davis, 4860 Y Street, Suite 3700, Sacramento, CA, 95817, USA; Center for Neuroscience, University of California, Davis, 1544 Newton Court, Davis, CA, 95618, USA.
| | - Randi H Hagerman
- MIND Institute, University of California, Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| | - John M Olichney
- Department of Neurology, University of California, Davis, 4860 Y Street, Suite 3700, Sacramento, CA, 95817, USA; Center for Mind and Brain, University of California, Davis, 267 Cousteau Place, Davis, CA, 95618, USA.
| |
Collapse
|
9
|
Cieślik P, Wierońska JM. Regulation of Glutamatergic Activity via Bidirectional Activation of Two Select Receptors as a Novel Approach in Antipsychotic Drug Discovery. Int J Mol Sci 2020; 21:ijms21228811. [PMID: 33233865 PMCID: PMC7699963 DOI: 10.3390/ijms21228811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/10/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022] Open
Abstract
Schizophrenia is a mental disorder that affects approximately 1-2% of the population and develops in early adulthood. The disease is characterized by positive, negative, and cognitive symptoms. A large percentage of patients with schizophrenia have a treatment-resistant disease, and the risk of developing adverse effects is high. Many researchers have attempted to introduce new antipsychotic drugs to the clinic, but most of these treatments failed, and the diversity of schizophrenic symptoms is one of the causes of disappointing results. The present review summarizes the results of our latest papers, showing that the simultaneous activation of two receptors with sub-effective doses of their ligands induces similar effects as the highest dose of each compound alone. The treatments were focused on inhibiting the increased glutamate release responsible for schizophrenia arousal, without interacting with dopamine (D2) receptors. Ligands activating metabotropic receptors for glutamate, GABAB or muscarinic receptors were used, and the compounds were administered in several different combinations. Some combinations reversed all schizophrenia-related deficits in animal models, but others were active only in select models of schizophrenia symptoms (i.e., cognitive or negative symptoms).
Collapse
|
10
|
Wang HY, MacDonald ML, Borgmann-Winter KE, Banerjee A, Sleiman P, Tom A, Khan A, Lee KC, Roussos P, Siegel SJ, Hemby SE, Bilker WB, Gur RE, Hahn CG. mGluR5 hypofunction is integral to glutamatergic dysregulation in schizophrenia. Mol Psychiatry 2020; 25:750-760. [PMID: 30214040 PMCID: PMC7500805 DOI: 10.1038/s41380-018-0234-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/18/2018] [Accepted: 07/09/2018] [Indexed: 11/19/2022]
Abstract
Multiple lines of evidence point to glutamatergic signaling in the postsynaptic density (PSD) as a pathophysiologic mechanism in schizophrenia. Integral to PSD glutamatergic signaling is reciprocal interplay between GluN and mGluR5 signaling. We examined agonist-induced mGluR5 signaling in the postmortem dorsolateral prefrontal cortex (DLPFC) derived from 17 patients and age-matched and sex-matched controls. The patient group showed a striking reduction in mGluR5 signaling, manifested by decreases in Gq/11 coupling and association with PI3K and Homer compared to controls (p < 0.01 for all). This was accompanied by increases in serine and tyrosine phosphorylation of mGluR5, which can decrease mGluR5 activity via desensitization (p < 0.01). In addition, we find altered protein-protein interaction (PPI) of mGluR5 with RGS4, norbin, Preso 1 and tamalin, which can also attenuate mGluR5 activity. We previously reported molecular underpinnings of GluN hypofunction (decreased GluN2 phosphorylation) and here we show those of reduced mGluR5 signaling in schizophrenia. We find that reduced GluN2 phosphorylation can be precipitated by attenuated mGluR5 activity and that increased mGluR5 phosphorylation can result from decreased GluN function, suggesting a reciprocal interplay between the two pathways in schizophrenia. Interestingly, the patient group showed decreased mGluR5-GluN association (p < 0.01), a mechanistic basis for the reciprocal facilitation. In sum, we present the first direct evidence for mGluR5 hypoactivity, propose a reciprocal interplay between GluN and mGluR5 pathways as integral to glutamatergic dysregulation and suggest protein-protein interactions in mGluR5-GluN complexes as potential targets for intervention in schizophrenia.
Collapse
Affiliation(s)
- Hoau-Yan Wang
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine, New York, NY 10031,Department of Biology and Neuroscience, Graduate School of the City University of New York, NY 10016
| | - Mathew L. MacDonald
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403
| | - Karin E. Borgmann-Winter
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403,Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Anamika Banerjee
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403
| | - Patrick Sleiman
- The Center for Applied Genomics, The Children’s Hospital of Philadelphia, and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104
| | - Andrew Tom
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine, New York, NY 10031
| | - Amber Khan
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine, New York, NY 10031,Department of Biology and Neuroscience, Graduate School of the City University of New York, NY 10016
| | - Kuo-Chieh Lee
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine, New York, NY 10031
| | - Panos Roussos
- Department of Psychiatry, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Steven J. Siegel
- Department of Psychiatry and the Behavioral Sciences, University of Southern California, Los Angeles, CA, 90007
| | - Scott E Hemby
- Department of Basic Pharmaceutical Sciences, High Point University, High Point, NC, 27106
| | - Warren B. Bilker
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA 19104
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104-3403
| | - Chang-Gyu Hahn
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104-3403, USA.
| |
Collapse
|
11
|
Dal Prà I, Armato U, Chiarini A. Family C G-Protein-Coupled Receptors in Alzheimer's Disease and Therapeutic Implications. Front Pharmacol 2019; 10:1282. [PMID: 31719824 PMCID: PMC6826475 DOI: 10.3389/fphar.2019.01282] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), particularly its sporadic or late-onset form (SAD/LOAD), is the most prevalent (96–98% of cases) neurodegenerative dementia in aged people. AD’s neuropathology hallmarks are intrabrain accumulation of amyloid-β peptides (Aβs) and of hyperphosphorylated Tau (p-Tau) proteins, diffuse neuroinflammation, and progressive death of neurons and oligodendrocytes. Mounting evidences suggest that family C G-protein-coupled receptors (GPCRs), which include γ-aminobutyric acid B receptors (GABABRs), metabotropic glutamate receptors (mGluR1-8), and the calcium-sensing receptor (CaSR), are involved in many neurotransmitter systems that dysfunction in AD. This review updates the available knowledge about the roles of GPCRs, particularly but not exclusively those expressed by brain astrocytes, in SAD/LOAD onset and progression, taking stock of their respective mechanisms of action and of their potential as anti-AD therapeutic targets. In particular, GABABRs prevent Aβs synthesis and neuronal hyperexcitability and group I mGluRs play important pathogenetic roles in transgenic AD-model animals. Moreover, the specific binding of Aβs to the CaSRs of human cortical astrocytes and neurons cultured in vitro engenders a pathological signaling that crucially promotes the surplus synthesis and release of Aβs and hyperphosphorylated Tau proteins, and also of nitric oxide, vascular endothelial growth factor-A, and proinflammatory agents. Concurrently, Aβs•CaSR signaling hinders the release of soluble (s)APP-α peptide, a neurotrophic agent and GABABR1a agonist. Altogether these effects progressively kill human cortical neurons in vitro and likely also in vivo. Several CaSR’s negative allosteric modulators suppress all the noxious effects elicited by Aβs•CaSR signaling in human cortical astrocytes and neurons thus safeguarding neurons’ viability in vitro and raising hopes about their potential therapeutic benefits in AD patients. Further basic and clinical investigations on these hot topics are needed taking always heed that activation of the several brain family C GPCRs may elicit divergent upshots according to the models studied.
Collapse
Affiliation(s)
- Ilaria Dal Prà
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Anna Chiarini
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| |
Collapse
|
12
|
Neyman S, Braunewell KH, O'Connell KE, Dev KK, Manahan-Vaughan D. Inhibition of the Interaction Between Group I Metabotropic Glutamate Receptors and PDZ-Domain Proteins Prevents Hippocampal Long-Term Depression, but Not Long-Term Potentiation. Front Synaptic Neurosci 2019; 11:13. [PMID: 31057390 PMCID: PMC6482240 DOI: 10.3389/fnsyn.2019.00013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/04/2019] [Indexed: 01/07/2023] Open
Abstract
The group I metabotropic glutamate (mGlu) receptor subtypes, mGlu1 and mGlu5, strongly regulate hippocampal synaptic plasticity. Both harbor PSD-95/discs-large/ZO-1 (PDZ) motifs at their extreme carboxyl terminals, which allow interaction with the PDZ domain of Tamalin, regulate the cell surface expression of group I mGlu receptors, and may modulate their coupling to signaling proteins. We investigated the functional role of this interaction in hippocampal long-term depression (LTD). Acute intracerebral treatment of adult rats with a cell-permeable PDZ-blocking peptide (pep-mGluR-STL), designed to competitively inhibit the interaction between Tamalin and group 1 mGlu receptors, prevented expression of LTD in the hippocampal CA1 region without affecting long-term potentiation (LTP) or basal synaptic transmission. Pep-mGluR-STL prevented facilitation by the group I mGlu receptor agonist, (S)-3,5-Dihydroxyphenylglycine (DHPG), and the mGlu5 agonist, (R,S)-2-chloro-5-Hydroxyphenylglycine (CHPG), of short-term depression (STD) into LTD, suggesting that Tamalin preferentially acts by mediating signaling through mGlu5. These data support that Tamalin is essential for the persistent expression of LTD and that it subserves the effective signaling of group 1 mGlu receptors.
Collapse
Affiliation(s)
- Sergey Neyman
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Karl-Heinz Braunewell
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Kara E O'Connell
- Drug Development, School of Medicine, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
13
|
Peñasco S, Rico-Barrio I, Puente N, Gómez-Urquijo SM, Fontaine CJ, Egaña-Huguet J, Achicallende S, Ramos A, Reguero L, Elezgarai I, Nahirney PC, Christie BR, Grandes P. Endocannabinoid long-term depression revealed at medial perforant path excitatory synapses in the dentate gyrus. Neuropharmacology 2019; 153:32-40. [PMID: 31022405 DOI: 10.1016/j.neuropharm.2019.04.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/28/2019] [Accepted: 04/18/2019] [Indexed: 12/22/2022]
Abstract
The endocannabinoid system modulates synaptic plasticity in the hippocampus, but a link between long-term synaptic plasticity and the type 1 cannabinoid (CB1) receptor at medial perforant path (MPP) synapses remains elusive. Here, immuno-electron microscopy in adult mice showed that ∼26% of the excitatory synaptic terminals in the middle 1/3 of the dentate molecular layer (DML) contained CB1 receptors, and field excitatory postsynaptic potentials evoked by MPP stimulation were inhibited by CB1 receptor activation. In addition, MPP stimulation at 10 Hz for 10 min triggered CB1 receptor-dependent excitatory long-term depression (eCB-eLTD) at MPP synapses of wild-type mice but not on CB1-knockout mice. This eCB-eLTD was group I mGluR-dependent, required intracellular calcium influx and 2-arachydonoyl-glycerol (2-AG) synthesis but did not depend on N-methyl-d-aspartate (NMDA) receptors. Overall, these results point to a functional role for CB1 receptors with eCB-eLTD at DML MPP synapses and further involve these receptors in memory processing within the adult brain.
Collapse
Affiliation(s)
- Sara Peñasco
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Irantzu Rico-Barrio
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Nagore Puente
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Sonia María Gómez-Urquijo
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Christine J Fontaine
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada
| | - Jon Egaña-Huguet
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Svein Achicallende
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Almudena Ramos
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Leire Reguero
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Izaskun Elezgarai
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Patrick C Nahirney
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada
| | - Pedro Grandes
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940, Leioa, Spain; Division of Medical Sciences, University of Victoria, Victoria, British Columbia, V8P 5C2, Canada.
| |
Collapse
|
14
|
Nicoletti F, Orlando R, Di Menna L, Cannella M, Notartomaso S, Mascio G, Iacovelli L, Matrisciano F, Fazio F, Caraci F, Copani A, Battaglia G, Bruno V. Targeting mGlu Receptors for Optimization of Antipsychotic Activity and Disease-Modifying Effect in Schizophrenia. Front Psychiatry 2019; 10:49. [PMID: 30890967 PMCID: PMC6413697 DOI: 10.3389/fpsyt.2019.00049] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/23/2019] [Indexed: 01/03/2023] Open
Abstract
Metabotropic glutamate (mGlu) receptors are considered as candidate drug targets for the treatment of schizophrenia. These receptors form a family of eight subtypes (mGlu1 to -8), of which mGlu1 and -5 are coupled to Gq/11, and all other subtypes are coupled to Gi/o. Here, we discuss the possibility that selective ligands of individual mGlu receptor subtypes may be effective in controlling the core symptoms of schizophrenia, and, in some cases, may impact mechanisms underlying the progression of the disorder. Recent evidence indicates that activation of mGlu1 receptors inhibits dopamine release in the meso-striatal system. Hence, selective positive allosteric modulators (PAMs) of mGlu1 receptors hold promise for the treatment of positive symptoms of schizophrenia. mGlu5 receptors are widely expressed in the CNS and regulate the activity of cells that are involved in the pathophysiology of schizophrenia, such as cortical GABAergic interneurons and microglial cells. mGlu5 receptor PAMs are under development for the treatment of schizophrenia and cater the potential to act as disease modifiers by restraining neuroinflammation. mGlu2 receptors have attracted considerable interest because they negatively modulate 5-HT2A serotonin receptor signaling in the cerebral cortex. Both mGlu2 receptor PAMs and orthosteric mGlu2/3 receptor agonists display antipsychotic-like activity in animal models, and the latter drugs are inactive in mice lacking mGlu2 receptors. So far, mGlu3 receptors have been left apart as drug targets for schizophrenia. However, activation of mGlu3 receptors boosts mGlu5 receptor signaling, supports neuronal survival, and drives microglial cells toward an antiinflammatory phenotype. This strongly encourages research of mGlu3 receptors in schizophrenia. Finally, preclical studies suggest that mGlu4 receptors might be targeted by novel antipsychotic drugs, whereas studies of mGlu7 and mGlu8 receptors in animal models of psychosis are still at their infancy.
Collapse
Affiliation(s)
- Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Rosamaria Orlando
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | - Luisa Iacovelli
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Francesco Matrisciano
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois, Chicago, IL, United States
| | | | - Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy.,Oasi Research Institute (IRCCS), Troina, Italy
| | - Agata Copani
- Department of Drug Sciences, University of Catania, Catania, Italy.,Institute of Biostructure and Bioimaging, National Research Council, Catania, Italy
| | | | - Valeria Bruno
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
15
|
Wierońska JM, Pilc A. Depression and schizophrenia viewed from the perspective of amino acidergic neurotransmission: Antipodes of psychiatric disorders. Pharmacol Ther 2018; 193:75-82. [PMID: 30149102 DOI: 10.1016/j.pharmthera.2018.08.010] [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: 01/21/2023]
Abstract
Depression and schizophrenia are burdensome, costly serious and disabling mental disorders. Moreover the existing treatments are not satisfactory. As amino-acidergic (AA) neurotransmitters built a vast majority of brain neurons, in this article we plan to focus on drugs influencing AA neurotransmission in both diseases: we will discuss several facts concerning glutamatergic and GABA-ergic neurotransmission in these diseases, based mainly on preclinical experiments that used stimulators and/or blockers of both neurotransmitter systems. In general a picture emerges showing, that treatments that increase excitatory effects (with either antagonists or agonists) tend to evoke antidepressant effects, while treatments that increase inhibitory effects tend to display antipsychotic properties. Moreover, it seems that the antidepressant activity of a given compound excludes it as a potential antipsychotic and vice versa.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland
| | - Andrzej Pilc
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland.
| |
Collapse
|
16
|
Zuena AR, Iacovelli L, Orlando R, Di Menna L, Casolini P, Alemà GS, Di Cicco G, Battaglia G, Nicoletti F. In Vivo Non-radioactive Assessment of mGlu5 Receptor-Activated Polyphosphoinositide Hydrolysis in Response to Systemic Administration of a Positive Allosteric Modulator. Front Pharmacol 2018; 9:804. [PMID: 30108503 PMCID: PMC6079191 DOI: 10.3389/fphar.2018.00804] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/03/2018] [Indexed: 01/10/2023] Open
Abstract
mGlu5 receptor-mediated polyphosphoinositide (PI) hydrolysis is classically measured by determining the amount of radioactivity incorporated in inositolmonophosphate (InsP) after labeling of membrane phospholipids with radioactive inositol. Although this method is historically linked to the study of mGlu receptors, it is inappropriate for the assessment of mGlu5 receptor signaling in vivo. Using a new ELISA kit we showed that systemic treatment with the selective positive allosteric modulator (PAM) of mGlu5 receptors VU0360172 enhanced InsP formation in different brain regions of CD1 or C57Black mice. The action of VU0360172 was sensitive to the mGlu5 receptor, negative allosteric modulator (NAM), MTEP, and was abolished in mice lacking mGlu5 receptors. In addition, we could demonstrate that endogenous activation of mGlu5 receptors largely accounted for the basal PI hydrolysis particularly in the prefrontal cortex. This method offers opportunity for investigation of mGlu5 receptor signaling in physiology and pathology, and could be used for the functional screening of mGlu5 receptor PAMs in living animals.
Collapse
Affiliation(s)
- Anna R Zuena
- Department of Physiology and Pharmacology "Vittorio Erspamer," Sapienza University, Rome, Italy
| | - Luisa Iacovelli
- Department of Physiology and Pharmacology "Vittorio Erspamer," Sapienza University, Rome, Italy
| | - Rosamaria Orlando
- Department of Physiology and Pharmacology "Vittorio Erspamer," Sapienza University, Rome, Italy
| | | | - Paola Casolini
- Department of Physiology and Pharmacology "Vittorio Erspamer," Sapienza University, Rome, Italy
| | | | - Gabriele Di Cicco
- Department of Physiology and Pharmacology "Vittorio Erspamer," Sapienza University, Rome, Italy
| | | | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology "Vittorio Erspamer," Sapienza University, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
17
|
Esterlis I, Holmes SE, Sharma P, Krystal JH, DeLorenzo C. Metabotropic Glutamatergic Receptor 5 and Stress Disorders: Knowledge Gained From Receptor Imaging Studies. Biol Psychiatry 2018; 84:95-105. [PMID: 29100629 PMCID: PMC5858955 DOI: 10.1016/j.biopsych.2017.08.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022]
Abstract
The metabotropic glutamatergic receptor subtype 5 (mGluR5) may represent a promising therapeutic target for stress-related psychiatric disorders. Here, we describe mGluR5 findings in stress disorders, particularly major depressive disorder (MDD), highlighting insights from positron emission tomography studies. Positron emission tomography studies report either no differences or lower mGluR5 in MDD, potentially reflecting MDD heterogeneity. Unlike the rapidly acting glutamatergic agent ketamine, mGluR5-specific modulation has not yet shown antidepressant efficacy in MDD and bipolar disorder. Although we recently showed that ketamine may work, in part, through significant mGluR5 modulation, the specific role of mGluR5 downregulation in ketamine's antidepressant response is unclear. In contrast to MDD, there has been much less investigation of mGluR5 in bipolar disorder, yet initial studies indicate that mGluR5-specific treatments may aid in both depressed and manic mood states. The direction of modulation needed may be state dependent, however, limiting clinical feasibility. There has been relatively little study of posttraumatic stress disorder or obsessive-compulsive disorder to date, although there is evidence for the upregulation of mGluR5 in these disorders. However, while antagonism of mGluR5 may reduce fear conditioning, it may also reduce fear extinction. Therefore, studies are needed to determine the role mGluR5 modulation might play in the treatment of these conditions. Further challenges in modulating this prevalent neurotransmitter system include potential induction of significant side effects. As such, more research is needed to identify level and type (positive/negative allosteric modulation or full antagonism) of mGluR5 modulation required to translate existing knowledge into improved therapies.
Collapse
Affiliation(s)
- Irina Esterlis
- Department of Psychiatry, Yale University, New Haven, Connecticut; US Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veteran's Affairs Connecticut Healthcare System, West Haven, Connecticut.
| | | | - Priya Sharma
- Department of Psychiatry, Schulich School of Medicine and Dentistry; Western University- London, Ontario, Canada; London Health Sciences Centre- Victoria Hospital
| | - John H. Krystal
- Yale University, Department of Psychiatry,Yale University, Department of Neuroscience,U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System
| | - Christine DeLorenzo
- Stony Brook University, Department of Psychiatry,Stony Brook University, Department of Biomedical Engineering
| |
Collapse
|
18
|
Barnes SA, Sheffler DJ, Semenova S, Cosford NDP, Bespalov A. Metabotropic Glutamate Receptor 5 as a Target for the Treatment of Depression and Smoking: Robust Preclinical Data but Inconclusive Clinical Efficacy. Biol Psychiatry 2018; 83:955-962. [PMID: 29628194 PMCID: PMC5953810 DOI: 10.1016/j.biopsych.2018.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022]
Abstract
The ability of novel pharmacological compounds to improve outcomes in preclinical models is often not translated into clinical efficacy. Psychiatric disorders do not have biological boundaries, and identifying mechanisms to improve the translational bottleneck between preclinical and clinical research domains is an important and challenging task. Glutamate transmission is disrupted in several neuropsychiatric disorders. Metabotropic glutamate (mGlu) receptors represent a diverse class of receptors that contribute to excitatory neurotransmission. Given the wide, yet region-specific manner of expression, developing pharmacological compounds to modulate mGlu receptor activity provides an opportunity to subtly and selectively modulate excitatory neurotransmission. This review focuses on the potential involvement of mGlu5 receptor disruption in major depressive disorder and substance and/or alcohol use disorders. We provide an overview of the justification of targeting mGlu5 receptors in the treatment of these disorders, summarize the preclinical evidence for negatively modulating mGlu5 receptors as a therapeutic target for major depressive disorders and nicotine dependence, and highlight the outcomes of recent clinical trials. While the evidence of mGlu5 receptor negative allosteric modulation has been promising in preclinical investigations, these beneficial effects have not translated into clinical efficacy. In this review, we identify key challenges that may contribute to poor clinical translation and provide suggested approaches moving forward to potentially improve the translation from preclinical to clinical domains. Such approaches may increase the success of clinical trials and may reduce the translational bottleneck that exists in drug discovery for psychiatric disorders.
Collapse
Affiliation(s)
- Samuel A. Barnes
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0603, La Jolla, CA 92093, USA
| | - Douglas J. Sheffler
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Svetlana Semenova
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0603, La Jolla, CA 92093, USA,PAREXEL International, 1560 E Chevy Chase Dr, suite 140, Glendale, CA 91206, USA
| | - Nicholas D. P. Cosford
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Anton Bespalov
- EXCIVA, Heidelberg, Germany; Valdman Institute of Pharmacology, Pavlov Medical University, St. Petersburg, Russia.
| |
Collapse
|
19
|
Lin T, Dang S, Su Q, Zhang H, Zhang J, Zhang L, Zhang X, Lu Y, Li H, Zhu Z. The Impact and Mechanism of Methylated Metabotropic Glutamate Receptors 1 and 5 in the Hippocampus on Depression-Like Behavior in Prenatal Stress Offspring Rats. J Clin Med 2018; 7:jcm7060117. [PMID: 29882864 PMCID: PMC6025529 DOI: 10.3390/jcm7060117] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/06/2018] [Accepted: 05/11/2018] [Indexed: 12/14/2022] Open
Abstract
An increasing number of epidemiological investigations and animal models research suggest that prenatal stress (PS) could cause depression-like behavior in the offspring, which is sex specific. However, the underlying mechanisms remain to be elucidated. This study is to investigate the promoter methylation of metabotropic glutamate receptor 1 (mGluR1) and metabotropic Glutamate Receptor 5 (mGluR5) gene modification on PS induced depression-like behavior in offspring rats (OR). PS models were established, with or without 5-aza-2′-deoxycytidine (5-azaD, decitabine) treatment. Animal behavior was assessed by the sucrose preference test (SPT), forced swimming test (FST), and open field test (OFT). The mRNA and protein expression levels of mGluR1 and mGluR5 in the hippocampus of offspring were detected with quantitative real-time PCR and Western blot analysis, respectively. The promoter methylation in the hippocampus of mGluR1 and mGluR5 OR were also analyzed. SPT showed significantly reduced sucrose preference in PS induced OR. FST showed significantly prolonged immobility time in PS induced OR. OFT showed significantly reduced central residence time in PS induced OR and no significantly influence in rearing as well as in frequency of micturition. Moreover, the mRNA, protein expression levels, and gene promoter methylation level of mGluR1 and mGluR5 in the hippocampus were significantly increased in the PS induced male OR, while no significantly influence in the PS induced female OR. Furthermore, the PS induced effects in male OR could be reversed by the microinjection of 5-azaD. In conclusion, our results showed that the promoter methylation of mGluR1 and mGluR5 gene modification is only involved in PS induced depression-like behavior in male OR in a sex-specific manner. These findings might contribute to the understanding of the disease pathogenesis and clinical treatment in future.
Collapse
Affiliation(s)
- Tianwei Lin
- Shaanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, China.
| | - Shaokang Dang
- Department of Pharmacology, College of Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Qian Su
- Department of Pharmacology, College of Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Huiping Zhang
- Department of Pharmacology, College of Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Junli Zhang
- Shaanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, China.
| | - Lin Zhang
- Shaanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, China.
| | - Xiaoxiao Zhang
- Shaanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, China.
| | - Yong Lu
- Department of Pharmacology, College of Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Hui Li
- Department of Pharmacology, College of Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Zhongliang Zhu
- Shaanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, China.
- Department of Pharmacology, College of Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
20
|
Benlloch JM, González AJ, Pani R, Preziosi E, Jackson C, Murphy J, Barberá J, Correcher C, Aussenhofer S, Gareis D, Visvikis D, Bert J, Langstrom B, Farde L, Toth M, Haggkvist J, Caixeta FV, Kullander K, Somlai-Schweiger I, Schwaiger M. The MINDVIEW project: First results. Eur Psychiatry 2018; 50:21-27. [PMID: 29398564 DOI: 10.1016/j.eurpsy.2018.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 01/01/2023] Open
Abstract
We present the first results of the MINDVIEW project. An innovative imaging system for the human brain examination, allowing simultaneous acquisition of PET/MRI images, has been designed and constructed. It consists of a high sensitivity and high resolution PET scanner integrated in a novel, head-dedicated, radio frequency coil for a 3T MRI scanner. Preliminary measurements from the PET scanner show sensitivity 3 times higher than state-of-the-art PET systems that will allow safe repeated studies on the same patient. The achieved spatial resolution, close to 1 mm, will enable differentiation of relevant brain structures for schizophrenia. A cost-effective and simple method of radiopharmaceutical production from 11C-carbon monoxide and a mini-clean room has been demonstrated. It has been shown that 11C-raclopride has higher binding potential in a new VAAT null mutant mouse model of schizophrenia compared to wild type control animals. A significant reduction in TSPO binding has been found in gray matter in a small sample of drug-naïve, first episode psychosis patients, suggesting a reduced number or an altered function of immune cells in brain at early stage schizophrenia.
Collapse
Affiliation(s)
- José M Benlloch
- Institute for Instrumentation in Molecular Imaging (I3 M), Universidad Politécnica de Valencia - CSIC, Valencia, Spain
| | - Antonio J González
- Institute for Instrumentation in Molecular Imaging (I3 M), Universidad Politécnica de Valencia - CSIC, Valencia, Spain.
| | - Roberto Pani
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - Enrico Preziosi
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | | | | | | | | | | | | | - Dimitris Visvikis
- INSERM, UMR1101, LaTIM, Université de Bretagne Occidentale, Brest, France
| | - Julien Bert
- INSERM, UMR1101, LaTIM, Université de Bretagne Occidentale, Brest, France
| | | | - Lars Farde
- Dept. of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden; Precision Medicine & Genomics, AstraZeneca, PET Science Center, Karolinska Institutet, Stockholm, Sweden
| | - Miklos Toth
- Dept. of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Jenny Haggkvist
- Dept. of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Fabio V Caixeta
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Klas Kullander
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | | | - Markus Schwaiger
- Technische Universität München, Dept. of Nuclear Medicine, Munich, Germany
| |
Collapse
|
21
|
Zurawek D, Salerno-Kochan A, Dziedzicka-Wasylewska M, Nikiforuk A, Kos T, Popik P. Changes in the expression of metabotropic glutamate receptor 5 (mGluR5) in a ketamine-based animal model of schizophrenia. Schizophr Res 2018; 192:423-430. [PMID: 28433499 DOI: 10.1016/j.schres.2017.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/30/2017] [Accepted: 04/07/2017] [Indexed: 12/26/2022]
Abstract
It has been shown that the metabotropic glutamate receptor subtype 5 (mGluR5) is functionally associated with the NMDA subtype of the glutamate receptor family (NMDA receptors). These two receptors colocalize in brain regions associated with schizophrenia. Although the role of the NMDA receptor in cognitive and negative symptoms of schizophrenia is well studied, information about the role of mGluR5 receptors in schizophrenia is sparse. In our work, we show that subchronic administration of ketamine, a well-studied, non-competitive antagonist of NMDA receptors, caused cognitive deficits in rats as shown by testing novel object recognition (NOR). Moreover, we reveal that subchronic administration of ketamine increased the mRNA and protein expression levels of mGluR5 receptors in regions CA1 and CA3 of the dorsal part of the hippocampus, both of which are strongly associated with the formation of visual memory, which is tested via NOR. We postulate that increased expression of mGluR5 receptors in the dorsal part of the hippocampus may reflect compensatory changes to imbalanced glutamate neurotransmission associated with the hypoactivation of NMDA receptors.
Collapse
Affiliation(s)
- Dariusz Zurawek
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 31-343 Krakow, Smetna Street 12, Poland.
| | - Anna Salerno-Kochan
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 31-343 Krakow, Smetna Street 12, Poland
| | - Marta Dziedzicka-Wasylewska
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 31-343 Krakow, Smetna Street 12, Poland
| | - Agnieszka Nikiforuk
- Institute of Pharmacology, Polish Academy of Sciences, Department of Behavioural Neuroscience and Drug Development, 31-343 Krakow, Smetna Street 12, Poland
| | - Tomasz Kos
- Institute of Pharmacology, Polish Academy of Sciences, Department of Behavioural Neuroscience and Drug Development, 31-343 Krakow, Smetna Street 12, Poland
| | - Piotr Popik
- Faculty of Health Sciences, Collegium Medicum, Jagiellonian University, Krakow, Poland
| |
Collapse
|
22
|
Foster DJ, Conn PJ. Allosteric Modulation of GPCRs: New Insights and Potential Utility for Treatment of Schizophrenia and Other CNS Disorders. Neuron 2017; 94:431-446. [PMID: 28472649 PMCID: PMC5482176 DOI: 10.1016/j.neuron.2017.03.016] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/02/2017] [Accepted: 03/09/2017] [Indexed: 01/11/2023]
Abstract
G-protein-coupled receptors (GPCRs) play critical roles in regulating brain function. Recent advances have greatly expanded our understanding of these receptors as complex signaling machines that can adopt numerous conformations and modulate multiple downstream signaling pathways. While agonists and antagonists have traditionally been pursued to target GPCRs, allosteric modulators provide several mechanistic advantages, including the ability to distinguish between closely related receptor subtypes. Recently, the discovery of allosteric ligands that confer bias and modulate some, but not all, of a given receptor's downstream signaling pathways can provide pharmacological modulation of brain circuitry with remarkable precision. In addition, allosteric modulators with unprecedented specificity have been developed that can differentiate between subpopulations of a given receptor subtype based on the receptor's dimerization state. These advances are not only providing insight into the biological roles of specific receptor populations, but hold great promise for treating numerous CNS disorders.
Collapse
Affiliation(s)
- Daniel J Foster
- 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.
| |
Collapse
|
23
|
Matosin N, Newell KA, Quidé Y, Andrews JL, Teroganova N, Green MJ, Fernandez F. Effects of common GRM5 genetic variants on cognition, hippocampal volume and mGluR5 protein levels in schizophrenia. Brain Imaging Behav 2017; 12:509-517. [DOI: 10.1007/s11682-017-9712-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
24
|
Bruno V, Caraci F, Copani A, Matrisciano F, Nicoletti F, Battaglia G. The impact of metabotropic glutamate receptors into active neurodegenerative processes: A "dark side" in the development of new symptomatic treatments for neurologic and psychiatric disorders. Neuropharmacology 2016; 115:180-192. [PMID: 27140693 DOI: 10.1016/j.neuropharm.2016.04.044] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/22/2016] [Accepted: 04/28/2016] [Indexed: 12/17/2022]
Abstract
Metabotropic glutamate (mGlu) receptor ligands are under clinical development for the treatment of CNS disorders with high social and economic burden, such as schizophrenia, major depressive disorder (MDD), and Parkinson's disease (PD), and are promising drug candidates for the treatment of Alzheimer's disease (AD). So far, clinical studies have shown symptomatic effects of mGlu receptor ligands, but it is unknown whether these drugs act as disease modifiers or, at the opposite end, they accelerate disease progression by enhancing neurodegeneration. This is a fundamental issue in the treatment of PD and AD, and is also an emerging theme in the treatment of schizophrenia and MDD, in which neurodegeneration is also present and contribute to disease progression. Moving from in vitro data and preclinical studies, we discuss the potential impact of drugs targeting mGlu2, mGlu3, mGlu4 and mGlu5 receptor ligands on active neurodegeneration associated with AD, PD, schizophrenia, and MDD. We wish to highlight that our final comments on the best drug candidates are not influenced by commercial interests or by previous or ongoing collaborations with drug companies. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
Affiliation(s)
- Valeria Bruno
- Department of Physiology and Pharmacology, University Sapienza, 00185 Rome, Italy; I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy.
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; I.R.C.C.S. Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, 94018 Troina, Italy
| | - Agata Copani
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; National Research Council, Institute of Biostructure and Bioimaging (IBB-CNR), 95126 Catania, Italy
| | - Francesco Matrisciano
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, USA
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University Sapienza, 00185 Rome, Italy; I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy
| | | |
Collapse
|
25
|
Lee JY, Choe ES, Yang CH, Choi KH, Cheong JH, Jang CG, Seo JW, Yoon SS. The mGluR5 antagonist MPEP suppresses the expression and reinstatement, but not the acquisition, of the ethanol-conditioned place preference in mice. Pharmacol Biochem Behav 2016; 140:33-8. [DOI: 10.1016/j.pbb.2015.10.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/05/2015] [Accepted: 10/23/2015] [Indexed: 01/29/2023]
|
26
|
Sarantis K, Tsiamaki E, Kouvaros S, Papatheodoropoulos C, Angelatou F. Adenosine A₂A receptors permit mGluR5-evoked tyrosine phosphorylation of NR2B (Tyr1472) in rat hippocampus: a possible key mechanism in NMDA receptor modulation. J Neurochem 2015; 135:714-26. [PMID: 26303340 DOI: 10.1111/jnc.13291] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 07/23/2015] [Accepted: 08/07/2015] [Indexed: 12/30/2022]
Abstract
A great body of evidence points toward a functional interaction between metabotropic glutamate 5 receptors (mGluR5) and NMDA receptors (NMDAR) that enhances synaptic plasticity and cognition. However, the molecular mechanism underlying this interaction remains unclear. Here, we show that co-activation of mGluR5 and NMDAR in hippocampal slices synergistically leads to a robust phosphorylation of NR2B (Tyr1472), which is Src kinase dependent and is enabled by endogenous adenosine acting on A2A receptors. As it is well known, NR2B (Tyr1472) phosphorylation anchors NR2B-containing NMDARs to the surface of post-synaptic membranes, preventing their internalization. This is supported by our electrophysiological experiments showing that co-activation of mGluR5 and NMDARs robustly enhances NMDAR-dependent neuronal excitability recorded in CA1 hippocampal region, which temporally coincides with the robust increase in NR2B (Tyr1472) phosphorylation, depends on Src kinases and is also permitted by A2A receptors. Thus, we strongly suggest that NR2B (Tyr1472) phosphorylation constitutes, at least to some extent, the molecular mechanism underlying the mGluR5-mediated enhancement of NMDAR-dependent responses, which is modulated by A2A receptors. A better understanding of the molecular basis of mGluR5/NMDAR interaction would elucidate their role in synaptic plasticity processes as well as in pathological conditions. We propose the following molecular mechanism by which metabotropic Glutamate Receptor 5 (mGluR5) potentiate ionotropic Glutamate N-Methyl-D-Aspartate Receptor (NMDAR) responses in rat hippocampus. Co-activation of mGLUR5/NMDAR activates Src kinases, leading to NR2B(Tyr1472) phosphorylation, which anchors NR2B-containing NMDAR to the plasma membrane, thus inducing a robust increase in the NMDA-dependent excitability. Interestingly, adenosine A2A receptors license the mGluR5-induced NR2B(Tyr1472) phosphorylation.
Collapse
Affiliation(s)
| | - Eirini Tsiamaki
- Physiology Department, Medical School, University of Patras, Patras, Greece
| | - Stylianos Kouvaros
- Physiology Department, Medical School, University of Patras, Patras, Greece
| | | | - Fevronia Angelatou
- Physiology Department, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
27
|
Jin DZ, Xue B, Mao LM, Wang JQ. Metabotropic glutamate receptor 5 upregulates surface NMDA receptor expression in striatal neurons via CaMKII. Brain Res 2015; 1624:414-423. [PMID: 26256252 DOI: 10.1016/j.brainres.2015.07.053] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 07/23/2015] [Accepted: 07/30/2015] [Indexed: 12/29/2022]
Abstract
Metabotropic and ionotropic glutamate receptors are closely clustered in postsynaptic membranes and are believed to interact actively with each other to control excitatory synaptic transmission. Metabotropic glutamate receptor 5 (mGluR5), for example, has been well documented to potentiate ionotropic NMDA receptor activity, although underlying mechanisms are poorly understood. In this study, we investigated the role of mGluR5 in regulating trafficking and subcellular distribution of NMDA receptors in adult rat striatal neurons. We found that the mGluR1/5 agonist DHPG concentration-dependently increased NMDA receptor GluN1 and GluN2B subunit expression in the surface membrane. Meanwhile, DHPG reduced GluN1 and GluN2B levels in the intracellular compartment. The effect of DHPG was blocked by an mGluR5 selective antagonist MTEP but not by an mGluR1 selective antagonist 3-MATIDA. Pretreatment with an inhibitor or a specific inhibitory peptide for synapse-enriched Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) also blocked the DHPG-stimulated redistribution of GluN1 and GluN2B. In addition, DHPG enhanced CaMKIIα activity and elevated GluN2B phosphorylation at a CaMKII-sensitive site (serine 1303). These results demonstrate that mGluR5 regulates trafficking of NMDA receptors in striatal neurons. Activation of mGluR5 appears to induce rapid trafficking of GluN1 and GluN2B to surface membranes through a signaling pathway involving CaMKII.
Collapse
Affiliation(s)
- Dao-Zhong Jin
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Bing Xue
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Li-Min Mao
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
28
|
Ghoshal A, Conn PJ. The hippocampo-prefrontal pathway: a possible therapeutic target for negative and cognitive symptoms of schizophrenia. FUTURE NEUROLOGY 2015; 10:115-128. [PMID: 25825588 DOI: 10.2217/fnl.14.63] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The hippocampo-prefrontal (H-PFC) pathway has been linked to cognitive and emotional disturbances in several psychiatric disorders including schizophrenia. Preclinical evidence from the NMDA receptor antagonism rodent model of schizophrenia shows severe pathology selective to the H-PFC pathway. It is speculated that there is an increased excitatory drive from the hippocampus to the prefrontal cortex due to dysfunctions in the H-PFC plasticity, which may serve as the basis for the behavioral consequences observed in this rodent model. Thus, the H-PFC pathway is currently emerging as a promising therapeutic target for the negative and cognitive symptom clusters of schizophrenia. Here, we have reviewed the physiological, pharmacological and functional characteristics of the H-PFC pathway and we propose that allosteric activation of glutamatergic and cholinergic neurotransmission can serve as a plausible therapeutic approach.
Collapse
Affiliation(s)
- Ayan Ghoshal
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232 0697, USA
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232 0697, USA
| |
Collapse
|
29
|
Nghia NA, Hirasawa T, Kasai H, Obata C, Moriishi K, Mochizuki K, Koizumi S, Kubota T. Long-term imipramine treatment increases N-methyl-d-aspartate receptor activity and expression via epigenetic mechanisms. Eur J Pharmacol 2015; 752:69-77. [PMID: 25701723 DOI: 10.1016/j.ejphar.2015.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 02/03/2015] [Accepted: 02/10/2015] [Indexed: 01/10/2023]
Abstract
Imipramine, a major antidepressant, is known to inhibit reuptake of serotonin and norepinephrine, which contributes to recovery from major depressive disorder. It has recently been reported that acute imipramine treatment inhibits N-methyl-d-aspartate (NMDA) receptor activity. However, the mechanisms underlying long-term effects of imipramine have not been identified. We tested these distinct effects in mouse cortical neurons and found that acute (30s) imipramine treatment decreased Ca(2+) influx through NMDA receptors, whereas long-term treatment (48h) increased Ca(2+) influx via the same receptors. Furthermore, long-term treatment increased NMDA receptor 2B (NR2B) subunit expression via epigenetic changes, including increased acetylation of histones H3K9 and H3K27 in the NR2B promoter and decreased activity of histone deacetylase 3 (HDAC3) and HDAC4. These results suggest that the long-term effects of imipramine on NMDA receptors are quite different from its acute effects. Furthermore, increased NR2B expression via epigenetic alterations might be a part of the mechanism responsible for this long-term effect.
Collapse
Affiliation(s)
- Nguyen An Nghia
- Department of Epigenetic Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Takae Hirasawa
- Department of Epigenetic Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan; Japan Science and Technology Agency (JST), CREST, 4-1-8 Honcho, Kawaguchi, Saitama, Japan.
| | - Hirotake Kasai
- Department of Microbiology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Chie Obata
- Department of Epigenetic Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan; Japan Science and Technology Agency (JST), CREST, 4-1-8 Honcho, Kawaguchi, Saitama, Japan
| | - Kohji Moriishi
- Department of Microbiology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Kazuki Mochizuki
- Faculty of Life and Environmental Sciences, University of Yamanashi, Takeda, Kofu, Yamanashi 409-8510, Japan
| | - Schuichi Koizumi
- Department of Pharmacology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Takeo Kubota
- Department of Epigenetic Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| |
Collapse
|
30
|
Nowak G, Pomierny-Chamioło L, Siwek A, Niedzielska E, Pomierny B, Pałucha-Poniewiera A, Pilc A. Prolonged administration of antidepressant drugs leads to increased binding of [3H]MPEP to mGlu5 receptors. Neuropharmacology 2014; 84:46-51. [DOI: 10.1016/j.neuropharm.2014.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/17/2014] [Accepted: 04/22/2014] [Indexed: 11/28/2022]
|
31
|
Zhang S, Manahan-Vaughan D. Place field stability requires the metabotropic glutamate receptor, mGlu5. Hippocampus 2014; 24:1330-40. [PMID: 24910241 PMCID: PMC4280887 DOI: 10.1002/hipo.22314] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2014] [Indexed: 01/13/2023]
Abstract
The metabotropic glutamate (mGlu) receptors are critically involved in enabling the persistency of forms of synaptic plasticity that are believed to underlie hippocampus-dependent memory. These receptors and in particular, mGlu5, are also required for hippocampus-dependent learning and memory. In the hippocampus, synaptic plasticity is one of the mechanisms by which spatial information may be represented. Another mechanism involves increased firing of place cells. Place cells increase their firing activity when an animal is in a specific spatial location. Inhibition of factors that are essential for synaptic plasticity, such as N-methyl-d-aspartate receptors or protein synthesis, also impair place cell activity. This raises the question as to whether mGlu receptors, that are so important for synaptic plasticity and spatial memory, are also important for place cell encoding. We examined location-dependent place cell firing i.e. place fields. We observed that antagonism of mGlu5, using 2-methyl-6-(phenylethynyl) pyridine (MPEP) had no effect on place field profiles in a familiar environment. However, in a novel environment mGlu5-antagonism affected long-term place field stability, reduced place cell firing and spatial information. These data strongly suggest a role for mGlu5 in the mechanisms underlying informational content and long-term stability of place fields, and add to evidence supporting the importance of these receptors for hippocampal function.
Collapse
Affiliation(s)
- Sijie Zhang
- Department of Neurophysiology, Medical Faculty; International Graduate School for Neuroscience, Ruhr University Bochum, Germany
| | | |
Collapse
|
32
|
Yin S, Niswender CM. Progress toward advanced understanding of metabotropic glutamate receptors: structure, signaling and therapeutic indications. Cell Signal 2014; 26:2284-97. [PMID: 24793301 DOI: 10.1016/j.cellsig.2014.04.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 04/27/2014] [Indexed: 12/24/2022]
Abstract
The metabotropic glutamate (mGlu) receptors are a group of Class C seven-transmembrane spanning/G protein-coupled receptors (7TMRs/GPCRs). These receptors are activated by glutamate, one of the standard amino acids and the major excitatory neurotransmitter. By activating G protein-dependent and non-G protein-dependent signaling pathways, mGlus modulate glutamatergic transmission both in the periphery and throughout the central nervous system. Since the discovery of the first mGlu receptor, and especially during the last decade, a great deal of progress has been made in understanding the signaling, structure, pharmacological manipulation and therapeutic indications of the 8 mGlu members.
Collapse
Affiliation(s)
- Shen Yin
- Department of Pharmacology, Vanderbilt University Medical School, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical School, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University Medical School, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical School, Nashville, TN 37232, USA.
| |
Collapse
|
33
|
D'Antoni S, Spatuzza M, Bonaccorso CM, Musumeci SA, Ciranna L, Nicoletti F, Huber KM, Catania MV. Dysregulation of group-I metabotropic glutamate (mGlu) receptor mediated signalling in disorders associated with Intellectual Disability and Autism. Neurosci Biobehav Rev 2014; 46 Pt 2:228-41. [PMID: 24548786 DOI: 10.1016/j.neubiorev.2014.02.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/13/2014] [Accepted: 02/06/2014] [Indexed: 12/11/2022]
Abstract
Activation of group-I metabotropic glutamate receptors, mGlu1 and mGlu5, triggers a variety of signalling pathways in neurons and glial cells, which are differently implicated in synaptic plasticity. The earliest and much of key studies discovered abnormal mGlu5 receptor function in Fragile X syndrome (FXS) mouse models which then motivated more recent work that finds mGlu5 receptor dysfunction in related disorders such as intellectual disability (ID), obsessive-compulsive disorder (OCD) and autism. Therefore, mGlu1/5 receptor dysfunction may represent a common aetiology of these complex diseases. Furthermore, many studies have focused on dysregulation of mGlu5 signalling to synaptic protein synthesis. However, emerging evidence finds abnormal mGlu5 receptor interactions with its scaffolding proteins in FXS which results in mGlu5 receptor dysfunction and phenotypes independent of signalling to protein synthesis. Finally, both an increased and reduced mGlu5 functioning seem to be associated with ID and autism spectrum disorders, with important consequences for potential treatment of these developmental disorders.
Collapse
Affiliation(s)
- Simona D'Antoni
- Institute of Neurological Sciences, the National Research Council of Italy (CNR), Catania, Italy
| | - Michela Spatuzza
- Institute of Neurological Sciences, the National Research Council of Italy (CNR), Catania, Italy
| | | | | | - Lucia Ciranna
- Department of Biomedical Sciences, section of Physiology, University of Catania, Italy
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli (IS), Italy; University of Rome La Sapienza, Rome, Italy
| | - Kimberly M Huber
- University of Texas Southwestern Medical Center, Department of Neuroscience, Dallas, TX 75390-9111, USA
| | - Maria Vincenza Catania
- Institute of Neurological Sciences, the National Research Council of Italy (CNR), Catania, Italy; IRCCS Oasi Maria SS, Troina (EN), Italy.
| |
Collapse
|
34
|
Newell KA, Matosin N. Rethinking metabotropic glutamate receptor 5 pathological findings in psychiatric disorders: implications for the future of novel therapeutics. BMC Psychiatry 2014; 14:23. [PMID: 24472577 PMCID: PMC3907147 DOI: 10.1186/1471-244x-14-23] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 01/21/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pharmacological modulation of metabotropic glutamate receptor 5 (mGluR5) is of marked interest as a novel therapeutic mechanism to treat schizophrenia and major depression. However, the status of mGluR5 in the pathophysiology of these disorders remains unknown. DISCUSSION The majority of studies in the schizophrenia post-mortem brain indicate that total mGluR5 expression is unaltered. However, close examination of the literature suggests that these findings are superficial, and in actuality, a number of critical factors have not yet been considered; alterations may be highly dependent on brain region, neuronal population or molecular organisation in specific cellular compartments. A number of genetic knockout studies (mGluR5, Norbin, Homer1 etc.) continue to lend support to a role of mGluR5 in the pathology of schizophrenia, providing impetus to explore the regulation of mGluR5 beyond total mGluR5 protein and mRNA levels. With regards to major depression, preliminary evidence to date shows a reduction in total mGluR5 protein and mRNA levels; however, as in schizophrenia, there are no studies examining mGluR5 function or regulation in the pathological state. A comprehensive understanding of mGluR5 regulation in major depression, particularly in comparison to schizophrenia, is crucial as this has extensive implications for mGluR5 targeting novel therapeutics, especially considering that opposing modulation of mGluR5 is of therapeutic interest for these two disorders. SUMMARY Despite the complexities, examinations of post-mortem human brain provide valuable insights into the pathologies of these inherently human disorders. It is important, especially with regards to the identification of novel therapeutic drug targets, to have an in depth understanding of the pathophysiologies of these disorders. We posit that brain region- and cell type-specific alterations exist in mGluR5 in schizophrenia and depression, with evidence pointing towards altered regulation of this receptor in psychiatric pathology. We consider the implications of these alterations, as well as the distinction between schizophrenia and depression, in the context of novel mGluR5 based therapeutics.
Collapse
Affiliation(s)
- Kelly A Newell
- Centre for Translational Neuroscience, Faculty of Science, Medicine and Health and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia.
| | - Natalie Matosin
- Centre for Translational Neuroscience, Faculty of Science, Medicine and Health and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia,Schizophrenia Research Institute, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
35
|
Hill WD, Davies G, van de Lagemaat LN, Christoforou A, Marioni RE, Fernandes CPD, Liewald DC, Croning MDR, Payton A, Craig LCA, Whalley LJ, Horan M, Ollier W, Hansell NK, Wright MJ, Martin NG, Montgomery GW, Steen VM, Le Hellard S, Espeseth T, Lundervold AJ, Reinvang I, Starr JM, Pendleton N, Grant SGN, Bates TC, Deary IJ. Human cognitive ability is influenced by genetic variation in components of postsynaptic signalling complexes assembled by NMDA receptors and MAGUK proteins. Transl Psychiatry 2014; 4:e341. [PMID: 24399044 PMCID: PMC3905224 DOI: 10.1038/tp.2013.114] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 09/12/2013] [Accepted: 10/21/2013] [Indexed: 12/11/2022] Open
Abstract
Differences in general cognitive ability (intelligence) account for approximately half of the variation in any large battery of cognitive tests and are predictive of important life events including health. Genome-wide analyses of common single-nucleotide polymorphisms indicate that they jointly tag between a quarter and a half of the variance in intelligence. However, no single polymorphism has been reliably associated with variation in intelligence. It remains possible that these many small effects might be aggregated in networks of functionally linked genes. Here, we tested a network of 1461 genes in the postsynaptic density and associated complexes for an enriched association with intelligence. These were ascertained in 3511 individuals (the Cognitive Ageing Genetics in England and Scotland (CAGES) consortium) phenotyped for general cognitive ability, fluid cognitive ability, crystallised cognitive ability, memory and speed of processing. By analysing the results of a genome wide association study (GWAS) using Gene Set Enrichment Analysis, a significant enrichment was found for fluid cognitive ability for the proteins found in the complexes of N-methyl-D-aspartate receptor complex; P=0.002. Replication was sought in two additional cohorts (N=670 and 2062). A meta-analytic P-value of 0.003 was found when these were combined with the CAGES consortium. The results suggest that genetic variation in the macromolecular machines formed by membrane-associated guanylate kinase (MAGUK) scaffold proteins and their interaction partners contributes to variation in intelligence.
Collapse
Affiliation(s)
- W D Hill
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - G Davies
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, UK,Medical Genetics Section, The University of Edinburgh Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, Western General Hospital Edinburgh, Edinburgh, UK
| | - L N van de Lagemaat
- Genes to Cognition Programme, Centre for Clinical Brain Sciences and Centre for Neuroregeneration The University of Edinburgh, Edinburgh, UK
| | - A Christoforou
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway,Dr E. Martens Research Group for Biological Psychiatry, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - R E Marioni
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, UK,Medical Genetics Section, The University of Edinburgh Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, Western General Hospital Edinburgh, Edinburgh, UK,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - C P D Fernandes
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway,Dr E. Martens Research Group for Biological Psychiatry, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - D C Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - M D R Croning
- Genes to Cognition Programme, Centre for Clinical Brain Sciences and Centre for Neuroregeneration The University of Edinburgh, Edinburgh, UK
| | - A Payton
- Centre for Integrated Genomic Medical Research, University of Manchester, Manchester, UK
| | - L C A Craig
- Public Health Nutrition Research Group Section of Population Health, University of Aberdeen, Aberdeen, UK
| | - L J Whalley
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - M Horan
- Centre for Clinical and Cognitive Neurosciences, Institute Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - W Ollier
- Centre for Integrated Genomic Medical Research, University of Manchester, Manchester, UK
| | - N K Hansell
- Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - M J Wright
- Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - N G Martin
- Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - G W Montgomery
- Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - V M Steen
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway,Dr E. Martens Research Group for Biological Psychiatry, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - S Le Hellard
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway,Dr E. Martens Research Group for Biological Psychiatry, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - T Espeseth
- Department of Psychology, University of Oslo, Oslo, Norway,KG Jebsen Centre for Psychosis Research, Oslo University Hospital, Oslo, Norway
| | - A J Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway,Kavli Research Centre for Aging and Dementia, Haraldplass Hospital, Bergen, Norway
| | - I Reinvang
- Department of Psychology, University of Oslo, Oslo, Norway
| | - J M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - N Pendleton
- Centre for Clinical and Cognitive Neurosciences, Institute Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - S G N Grant
- Genes to Cognition Programme, Centre for Clinical Brain Sciences and Centre for Neuroregeneration The University of Edinburgh, Edinburgh, UK
| | - T C Bates
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - I J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, UK,Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh EH8 9JZ, UK. E-mail:
| |
Collapse
|
36
|
Jin DZ, Guo ML, Xue B, Mao LM, Wang JQ. Differential regulation of CaMKIIα interactions with mGluR5 and NMDA receptors by Ca(2+) in neurons. J Neurochem 2013; 127:620-31. [PMID: 24032403 DOI: 10.1111/jnc.12434] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/05/2013] [Accepted: 08/21/2013] [Indexed: 11/29/2022]
Abstract
Two glutamate receptors, metabotropic glutamate receptor 5 (mGluR5), and ionotropic NMDA receptors (NMDAR), functionally interact with each other to regulate excitatory synaptic transmission in the mammalian brain. In exploring molecular mechanisms underlying their interactions, we found that Ca(2+) /calmodulin-dependent protein kinase IIα (CaMKIIα) may play a central role. The synapse-enriched CaMKIIα directly binds to the proximal region of intracellular C terminal tails of mGluR5 in vitro. This binding is state-dependent: inactive CaMKIIα binds to mGluR5 at a high level whereas the active form of the kinase (following Ca(2+) /calmodulin binding and activation) loses its affinity for the receptor. Ca(2+) also promotes calmodulin to bind to mGluR5 at a region overlapping with the CaMKIIα-binding site, resulting in a competitive inhibition of CaMKIIα binding to mGluR5. In rat striatal neurons, inactive CaMKIIα constitutively binds to mGluR5. Activation of mGluR5 Ca(2+) -dependently dissociates CaMKIIα from the receptor and simultaneously promotes CaMKIIα to bind to the adjacent NMDAR GluN2B subunit, which enables CaMKIIα to phosphorylate GluN2B at a CaMKIIα-sensitive site. Together, the long intracellular C-terminal tail of mGluR5 seems to serve as a scaffolding domain to recruit and store CaMKIIα within synapses. The mGluR5-dependent Ca(2+) transients differentially regulate CaMKIIα interactions with mGluR5 and GluN2B in striatal neurons, which may contribute to cross-talk between the two receptors. We show that activation of mGluR5 with a selective agonist triggers intracellular Ca(2+) release in striatal neurons. Released Ca(2+) dissociates preformed CaMKIIα from mGluR5 and meanwhile promotes active CaMKIIα to bind to the adjacent NMDAR GluN2B subunit, which enables CaMKIIα to phosphorylate GluN2B at a CaMKIIα-sensitive site. This agonist-induced cascade seems to mediate crosstalk between mGluR5 and NMDA receptors in neurons.
Collapse
Affiliation(s)
- Dao-Zhong Jin
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | | | | | | | | |
Collapse
|
37
|
Won H, Mah W, Kim E. Autism spectrum disorder causes, mechanisms, and treatments: focus on neuronal synapses. Front Mol Neurosci 2013; 6:19. [PMID: 23935565 PMCID: PMC3733014 DOI: 10.3389/fnmol.2013.00019] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/16/2013] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of developmental disabilities characterized by impairments in social interaction and communication and restricted and repetitive interests/behaviors. Advances in human genomics have identified a large number of genetic variations associated with ASD. These associations are being rapidly verified by a growing number of studies using a variety of approaches, including mouse genetics. These studies have also identified key mechanisms underlying the pathogenesis of ASD, many of which involve synaptic dysfunctions, and have investigated novel, mechanism-based therapeutic strategies. This review will try to integrate these three key aspects of ASD research: human genetics, animal models, and potential treatments. Continued efforts in this direction should ultimately reveal core mechanisms that account for a larger fraction of ASD cases and identify neural mechanisms associated with specific ASD symptoms, providing important clues to efficient ASD treatment.
Collapse
Affiliation(s)
- Hyejung Won
- Department of Biological Sciences, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
| | - Won Mah
- Department of Biological Sciences, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic ScienceDaejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic ScienceDaejeon, South Korea
| |
Collapse
|
38
|
Wierońska JM, Pilc A. Glutamate-based anxiolytic ligands in clinical trials. Expert Opin Investig Drugs 2013; 22:1007-22. [PMID: 23718208 DOI: 10.1517/13543784.2013.803066] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION With regard to anxiety, the role of the balance between glutamatergic and GABAergic systems was pursued for many years. The majority of drugs used presently as effective anxiolytics enhance the GABAergic system activity, thus increasing inhibition within the central nervous system (CNS). On the other hand, decreasing the activity of glutamatergic neurotransmission may attenuate excitation in the CNS, thus resulting in anxiolysis. AREAS COVERED The present review focuses on clinical data of well-known and recently discovered glutamatergic and, to a lesser extent, GABAergic agents, which reached at least the Phase II criteria. EXPERT OPINION A variety of glutamatergic agents active at both N-acetylo-D-asparaginian and metabotropic glutamate (mGlu) receptors have been tested in humans to examine their potential anxiolytic activity. Many compounds acting on the glutamatergic system and approved for the treatment of other disorders than anxiety were shown to exert anxiolytic effects in clinical trials. Those are mainly voltage-dependent ion channel ligands as well as d-cycloserin and memantine. Also, ligands active at mGlu receptors, such as fenobam and LY354740, exhibited activity in controlled clinical trials. However, relatively few trials are found on the agents that are focused on GABAergic neurotransmission. Therefore, it seems that glutamatergic system may become a novel target for modern and effective anxiolytics.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
| | | |
Collapse
|
39
|
Mouri A, Nagai T, Ibi D, Yamada K. Animal models of schizophrenia for molecular and pharmacological intervention and potential candidate molecules. Neurobiol Dis 2013; 53:61-74. [DOI: 10.1016/j.nbd.2012.10.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 10/23/2012] [Accepted: 10/28/2012] [Indexed: 12/22/2022] Open
|
40
|
Mukherjee S, Manahan-Vaughan D. Role of metabotropic glutamate receptors in persistent forms of hippocampal plasticity and learning. Neuropharmacology 2013; 66:65-81. [DOI: 10.1016/j.neuropharm.2012.06.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 05/31/2012] [Accepted: 06/01/2012] [Indexed: 12/27/2022]
|
41
|
In vitro characterisation of the novel positive allosteric modulators of the mGlu5 receptor, LSN2463359 and LSN2814617, and their effects on sleep architecture and operant responding in the rat. Neuropharmacology 2013; 64:224-39. [DOI: 10.1016/j.neuropharm.2012.07.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/14/2012] [Accepted: 07/16/2012] [Indexed: 11/18/2022]
|
42
|
Pharmacology of metabotropic glutamate receptor allosteric modulators: structural basis and therapeutic potential for CNS disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 115:61-121. [PMID: 23415092 DOI: 10.1016/b978-0-12-394587-7.00002-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The metabotropic glutamate receptors (mGlus) mediate a neuromodulatory role throughout the brain for the major excitatory neurotransmitter, glutamate. Seven of the eight mGlu subtypes are expressed within the CNS and are attractive targets for a variety of psychiatric and neurological disorders including anxiety, depression, schizophrenia, Parkinson's disease, and Fragile X syndrome. Allosteric modulation of these class C 7-transmembrane spanning receptors represents a novel approach to facilitate development of mGlu subtype-selective probes and therapeutics. Allosteric modulators that interact with sites topographically distinct from the endogenous ligand-binding site offer a number of advantages over their competitive counterparts. In particular for CNS therapeutics, allosteric modulators have the potential to maintain the spatial and temporal aspects of endogenous neurotransmission. The past 15 years have seen the discovery of numerous subtype-selective allosteric modulators for the majority of the mGlu family members, including positive, negative, and neutral allosteric modulators, with a number of mGlu allosteric modulators now in clinical trials.
Collapse
|
43
|
Matosin N, Newell KA. Metabotropic glutamate receptor 5 in the pathology and treatment of schizophrenia. Neurosci Biobehav Rev 2012; 37:256-68. [PMID: 23253944 DOI: 10.1016/j.neubiorev.2012.12.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 10/27/2012] [Accepted: 12/09/2012] [Indexed: 02/07/2023]
Abstract
Metabotropic glutamate receptor 5 (mGluR5) potentiates the NMDA receptor (NMDAR) in brain regions implicated in schizophrenia, making it a viable therapeutic target for the treatment of this disorder. mGluR5 positive allosteric modulators may represent a valuable novel strategy for schizophrenia treatment, given the favourable profile of effects in preclinical paradigms. However it remains unclear whether mGluR5 also plays a causal or epiphenomenal role in NMDAR dysfunction in schizophrenia. Animal and cellular data suggest involvement of mGluR5, whilst post-mortem human studies remain inconclusive. This review will explore the molecular, animal and human data to support and refute the involvement of mGluR5 in the pathology of schizophrenia. Furthermore, this review will discuss the potential of mGluR5 modulators in the therapy of schizophrenia as well as aspects of mGluR5 that require further characterisation.
Collapse
Affiliation(s)
- Natalie Matosin
- Centre for Translational Neuroscience, Illawarra Health and Medical Research Institute, School of Health Sciences, University of Wollongong, NSW 2522, Australia
| | | |
Collapse
|
44
|
Huang CC, Hsu KS. Activation of NMDA receptors reduces metabotropic glutamate receptor-induced long-term depression in the nucleus accumbens via a CaMKII-dependent mechanism. Neuropharmacology 2012; 63:1298-307. [DOI: 10.1016/j.neuropharm.2012.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 07/30/2012] [Accepted: 08/14/2012] [Indexed: 01/12/2023]
|
45
|
Piers TM, Kim DH, Kim BC, Regan P, Whitcomb DJ, Cho K. Translational Concepts of mGluR5 in Synaptic Diseases of the Brain. Front Pharmacol 2012. [PMID: 23205012 PMCID: PMC3506921 DOI: 10.3389/fphar.2012.00199] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The G-protein coupled receptor family of glutamate receptors, termed metabotropic glutamate receptors (mGluRs), are implicated in numerous cellular mechanisms ranging from neural development to the processing of cognitive, sensory, and motor information. Over the last decade, multiple mGluR-related signal cascades have been identified at excitatory synapses, indicating their potential roles in various forms of synaptic function and dysfunction. This review highlights recent studies investigating mGluR5, a subtype of group I mGluRs, and its association with a number of developmental, psychiatric, and senile synaptic disorders with respect to associated synaptic proteins, with an emphasis on translational pre-clinical studies targeting mGluR5 in a range of synaptic diseases of the brain.
Collapse
Affiliation(s)
- Thomas M Piers
- School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol Bristol, UK
| | | | | | | | | | | |
Collapse
|
46
|
Fowler SW, Walker JM, Klakotskaia D, Will MJ, Serfozo P, Simonyi A, Schachtman TR. Effects of a metabotropic glutamate receptor 5 positive allosteric modulator, CDPPB, on spatial learning task performance in rodents. Neurobiol Learn Mem 2012; 99:25-31. [PMID: 23137441 DOI: 10.1016/j.nlm.2012.10.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 10/10/2012] [Accepted: 10/23/2012] [Indexed: 11/28/2022]
Abstract
Metabotropic glutamate receptor 5 (mGlu5) has been implicated in a variety of learning and memory processes and is important for avoidance learning. The present studies used an mGlu5 receptor positive allosteric modulator, 3-cyano-N-(1,3 diphenyl-1H-hyrazol-5-yl)benzamide (CDPPB), to characterize the importance of mGlu5 receptors in aversively- and appetitively-motivated spatial learning tasks (tasks in which the instrumental contingency involves discriminative cues that differ in spatial location). C57Bl/6 male mice were initially trained in the Barnes maze in the absence of drug. Subsequently, CDPPB (30mg/kg, i.p.), administered 20min prior to each of 3 daily reversal learning training sessions in the Barnes maze, significantly enhanced performance compared to vehicle-treated controls and had a significant effect on search strategy. Mice treated with CDPPB also displayed significantly less perseverative behavior than control-treated animals. In a second experiment, male Sprague-Dawley rats were trained in an appetitively-motivated, delayed alternation version of a T-maze. 30mg/kg CDPPB (s.c.), delivered 20min prior to each of 5 daily training sessions, enhanced the delay rats were able to withstand between the sample and choice portions of each T-maze trial. The present results emphasize the role of mGlu5 receptors in spatial learning tasks and support previous studies which report mGlu5 positive allosteric modulators can enhance learning in some tasks and may have potential as nootropic drugs.
Collapse
Affiliation(s)
- S W Fowler
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, United States
| | | | | | | | | | | | | |
Collapse
|
47
|
Pałucha-Poniewiera A, Pilc A. Involvement of mGlu5 and NMDA receptors in the antidepressant-like effect of acamprosate in the tail suspension test. Prog Neuropsychopharmacol Biol Psychiatry 2012; 39:102-6. [PMID: 22651960 DOI: 10.1016/j.pnpbp.2012.05.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 05/08/2012] [Accepted: 05/22/2012] [Indexed: 02/02/2023]
Abstract
Accumulating evidence supports the hypothesis that modulation of glutamatergic system via NMDA receptors and mGlu5 receptors might be an effective antidepressant therapy. However, clinical application of NMDA and mGlu5 antagonists in the therapy of depression is still an open question. In the present study we investigated potential antidepressant-like effect of a functional NMDA and mGlu5 receptor antagonist, acamprosate, which has been used in the therapy of human alcoholics as an anti-craving drug for more than 20 years and is considered as a safe substance. We have found potential antidepressant-like effect of acamprosate at doses of 100-400 mg/kg in the TST in C57BL/6J mice. Furthermore we have shown that the antidepressant-like effect of acamprosate used at a dose of 200 mg/kg was dependent on NMDA and mGlu5 receptor blockade, since NMDA (25 mg/kg) and mGlu5 receptor positive allosteric modulator, CDPPB (3 mg/kg), antagonized its activity in the TST. These data suggest that acamprosate may induce antidepressant-like effect and that NMDA and mGlu5 receptors are crucial targets of acamprosate in this action.
Collapse
|
48
|
Hovelsø N, Sotty F, Montezinho LP, Pinheiro PS, Herrik KF, Mørk A. Therapeutic potential of metabotropic glutamate receptor modulators. Curr Neuropharmacol 2012; 10:12-48. [PMID: 22942876 PMCID: PMC3286844 DOI: 10.2174/157015912799362805] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Revised: 01/10/2011] [Accepted: 03/04/2011] [Indexed: 12/21/2022] Open
Abstract
Glutamate is the main excitatory neurotransmitter in the central nervous system (CNS) and is a major player in complex brain functions. Glutamatergic transmission is primarily mediated by ionotropic glutamate receptors, which include NMDA, AMPA and kainate receptors. However, glutamate exerts modulatory actions through a family of metabotropic G-protein-coupled glutamate receptors (mGluRs). Dysfunctions of glutamatergic neurotransmission have been implicated in the etiology of several diseases. Therefore, pharmacological modulation of ionotropic glutamate receptors has been widely investigated as a potential therapeutic strategy for the treatment of several disorders associated with glutamatergic dysfunction. However, blockade of ionotropic glutamate receptors might be accompanied by severe side effects due to their vital role in many important physiological functions. A different strategy aimed at pharmacologically interfering with mGluR function has recently gained interest. Many subtype selective agonists and antagonists have been identified and widely used in preclinical studies as an attempt to elucidate the role of specific mGluRs subtypes in glutamatergic transmission. These studies have allowed linkage between specific subtypes and various physiological functions and more importantly to pathological states. This article reviews the currently available knowledge regarding the therapeutic potential of targeting mGluRs in the treatment of several CNS disorders, including schizophrenia, addiction, major depressive disorder and anxiety, Fragile X Syndrome, Parkinson’s disease, Alzheimer’s disease and pain.
Collapse
Affiliation(s)
- N Hovelsø
- Department of Neurophysiology, H. Lundbeck A/S, Ottiliavej 9, 2500 Copenhagen-Valby, Denmark
| | | | | | | | | | | |
Collapse
|
49
|
Liu CY, Jiang XX, Zhu YH, Wei DN. Metabotropic glutamate receptor 5 antagonist 2-methyl-6-(phenylethynyl)pyridine produces antidepressant effects in rats: role of brain-derived neurotrophic factor. Neuroscience 2012; 223:219-24. [PMID: 22890078 DOI: 10.1016/j.neuroscience.2012.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Revised: 07/17/2012] [Accepted: 08/06/2012] [Indexed: 02/07/2023]
Abstract
Recent studies highlight that the brain glutamate system is involved in the etiology of depression and glutamatergic-targeting drugs are currently being explored as novel antidepressant medications. Previous studies reveal that the selective metabotropic glutamate receptor 5 antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP) produces antidepressant-like effects in behavioral despair and olfactory bulbectomy models. The current study aimed to further explore its behavioral actions in additional animal models of depression (forced swimming test (FST) and learned helplessness (LH) test) and its underlying neurobiological mechanisms. The results demonstrated that acute treatment of MPEP at 30 but not 10mg/kg significantly reduced immobility in FST without affecting locomotor activities. Sub-chronic, five-day treatment of MPEP (30 mg/kg) decreased escape failures in animals that had developed LH symptoms. This sub-chronic treatment also increased hippocampal brain-derived neurotrophic factor (BDNF) protein levels in both non-stressed and stressed animals and restored the stress-induced down-regulation of BDNF expression. Current findings provide strong evidence for further studies of MPEP as a tool to explore novel antidepressants.
Collapse
Affiliation(s)
- C Y Liu
- Department of Neurosurgery, The Third Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | | | | | | |
Collapse
|
50
|
Gastambide F, Gilmour G, Robbins TW, Tricklebank MD. The mGlu₅ positive allosteric modulator LSN2463359 differentially modulates motor, instrumental and cognitive effects of NMDA receptor antagonists in the rat. Neuropharmacology 2012; 64:240-7. [PMID: 22884612 DOI: 10.1016/j.neuropharm.2012.07.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/14/2012] [Accepted: 07/16/2012] [Indexed: 02/04/2023]
Abstract
Metabotropic glutamate 5 (mGlu₅) receptors are known to functionally interact with N-methyl-d-aspartate (NMDA) receptors at both neuronal and behavioural levels, in a manner that may be of relevance to the treatment of schizophrenia. We have previously described a novel mGlu₅ positive allosteric modulator (PAM), LSN2463359 and provided evidence of its ability to attenuate aspects of the behavioural response to administration of the competitive NMDA receptor antagonist, SDZ 220,581. In addition, LSN2463359 was found to selectively attenuate reversal learning deficits observed in the neurodevelopmental MAM E17 model but not in the acute phencyclidine (PCP) model. In the present study, the interactions between this mGlu₅ PAM and the NMDA receptor were explored further by assessing the effects of LSN2463359 against some of the motor, instrumental and cognitive effects induced by the non-competitive NMDA receptor antagonists PCP and MK-801, the competitive NMDA receptor antagonist SDZ 220,581 and the GluN2B selective NMDA receptor antagonist, Ro 63-1908. LSN2463359 had either no or minor impact on locomotor hyperactivity induced by either PCP or SDZ 220,581. However, in rats lever pressing for food rewards under a variable interval 30s schedule of instrumental responding, the drug clearly attenuated not only the suppression of response rate induced by SDZ 220,581 but also the stimulation of response rate induced by Ro 63-1908. In contrast, LSN2463359 failed to alter both of the instrumental effects induced by the open channel blockers PCP and MK-801. In addition, although PCP and SDZ 220,581 induced similar deficits in a discrimination and reversal learning task, LSN2463359 was again only able to reverse the deficit induced by SDZ 220,581. The results indicate that the interactions between mGlu₅ and NMDA receptors are dependent on both the mechanism of the blockade of the receptor and the behavioural domain under investigation. Our work has implications for the preclinical use of NMDA receptor antagonists in the prediction of potential therapeutic efficacy in the search for novel treatments for schizophrenia. Positive allosteric modulators of the mGlu₅ receptor certainly question the predictive validity of such approaches. This article is part of a Special Issue entitled 'Cognitive Enhancers'.
Collapse
Affiliation(s)
- Francois Gastambide
- Lilly Centre for Cognitive Neuroscience, Lilly Research Laboratories, Eli Lilly & Co. Ltd., Erl Wood Manor, Windlesham, Surrey, UK.
| | | | | | | |
Collapse
|