1
|
Parent HH, Niswender CM. Therapeutic Potential for Metabotropic Glutamate Receptor 7 Modulators in Cognitive Disorders. Mol Pharmacol 2024; 105:348-358. [PMID: 38423750 PMCID: PMC11026152 DOI: 10.1124/molpharm.124.000874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is the most highly conserved and abundantly expressed mGlu receptor in the human brain. The presynaptic localization of mGlu7, coupled with its low affinity for its endogenous agonist, glutamate, are features that contribute to the receptor's role in modulating neuronal excitation and inhibition patterns, including long-term potentiation, in various brain regions. These characteristics suggest that mGlu7 modulation may serve as a novel therapeutic strategy in disorders of cognitive dysfunction, including neurodevelopmental disorders that cause impairments in learning, memory, and attention. Primary mutations in the GRM7 gene have recently been identified as novel causes of neurodevelopmental disorders, and these patients exhibit profound intellectual and cognitive disability. Pharmacological tools, such as agonists, antagonists, and allosteric modulators, have been the mainstay for targeting mGlu7 in its endogenous homodimeric form to probe effects of its function and modulation in disease models. However, recent research has identified diversity in dimerization, as well as trans-synaptic interacting proteins, that also play a role in mGlu7 signaling and pharmacological properties. These novel findings represent exciting opportunities in the field of mGlu receptor drug discovery and highlight the importance of further understanding the functions of mGlu7 in complex neurologic conditions at both the molecular and physiologic levels. SIGNIFICANCE STATEMENT: Proper expression and function of mGlu7 is essential for learning, attention, and memory formation at the molecular level within neural circuits. The pharmacological targeting of mGlu7 is undergoing a paradigm shift by incorporating an understanding of receptor interaction with other cis- and trans- acting synaptic proteins, as well as various intracellular signaling pathways. Based upon these new findings, mGlu7's potential as a drug target in the treatment of cognitive disorders and learning impairments is primed for exploration.
Collapse
Affiliation(s)
- Harrison H Parent
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| | - Colleen M Niswender
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| |
Collapse
|
2
|
Freitas GA, Niswender CM. GRM7 gene mutations and consequences for neurodevelopment. Pharmacol Biochem Behav 2023; 225:173546. [PMID: 37003303 PMCID: PMC10192299 DOI: 10.1016/j.pbb.2023.173546] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
The metabotropic glutamate receptor 7 (mGlu7), encoded by the GRM7 gene in humans, is a presynaptic, G protein-coupled glutamate receptor that is essential for modulating neurotransmission. Mutations in or reduced expression of GRM7 have been identified in different genetic neurodevelopmental disorders (NDDs), and rare biallelic missense variants have been proposed to underlie a subset of NDDs. Clinical GRM7 variants have been associated with a range of symptoms consistent with neurodevelopmental molecular features, including hypomyelination, brain atrophy and defects in axon outgrowth. Here, we review the newest findings regarding the cellular and molecular defects caused by GRM7 variants in NDD patients.
Collapse
Affiliation(s)
- Geanne A Freitas
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37212, United States of America
| | - Colleen M Niswender
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37212, United States of America; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37212, United States of America; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37212, United States of America; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America.
| |
Collapse
|
3
|
Dickson L, Teall M, Chevalier E, Cheung T, Liwicki GM, Mack S, Stephenson A, White K, Fosbeary R, Harrison DC, Brice NL, Doyle K, Ciccocioppo R, Wu C, Almond S, Patel TR, Mitchell P, Barnes M, Ayscough AP, Dawson LA, Carlton M, Bürli RW. Discovery of CVN636: A Highly Potent, Selective, and CNS Penetrant mGluR 7 Allosteric Agonist. ACS Med Chem Lett 2023; 14:442-449. [PMID: 37077399 PMCID: PMC10107911 DOI: 10.1021/acsmedchemlett.2c00529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
The low affinity metabotropic glutamate receptor mGluR7 has been implicated in numerous CNS disorders; however, a paucity of potent and selective activators has hampered full delineation of the functional role and therapeutic potential of this receptor. In this work, we present the identification, optimization, and characterization of highly potent, novel mGluR7 agonists. Of particular interest is the chromane CVN636, a potent (EC50 7 nM) allosteric agonist which demonstrates exquisite selectivity for mGluR7 compared to not only other mGluRs, but also a broad range of targets. CVN636 demonstrated CNS penetrance and efficacy in an in vivo rodent model of alcohol use disorder. CVN636 thus has potential to progress as a drug candidate in CNS disorders involving mGluR7 and glutamatergic dysfunction.
Collapse
Affiliation(s)
- Louise Dickson
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Martin Teall
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Elodie Chevalier
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Toni Cheung
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Gemma M. Liwicki
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Stephen Mack
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Anne Stephenson
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Kathryn White
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Richard Fosbeary
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - David C. Harrison
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Nicola L. Brice
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Kevin Doyle
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
| | - Chaobo Wu
- WuXi Apptec Limited, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Sarah Almond
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Toshal R. Patel
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Philip Mitchell
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Matt Barnes
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Andrew P. Ayscough
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Lee A. Dawson
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Mark Carlton
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
- Takeda Cambridge Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| | - Roland W. Bürli
- Cerevance Limited, 418 Cambridge Science Park, Cambridge CB4 0PZ, U.K
| |
Collapse
|
4
|
Lukacs IP, Francavilla R, Field M, Hunter E, Howarth M, Horie S, Plaha P, Stacey R, Livermore L, Ansorge O, Tamas G, Somogyi P. Differential effects of group III metabotropic glutamate receptors on spontaneous inhibitory synaptic currents in spine-innervating double bouquet and parvalbumin-expressing dendrite-targeting GABAergic interneurons in human neocortex. Cereb Cortex 2023; 33:2101-2142. [PMID: 35667019 PMCID: PMC9977385 DOI: 10.1093/cercor/bhac195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/12/2022] Open
Abstract
Diverse neocortical GABAergic neurons specialize in synaptic targeting and their effects are modulated by presynaptic metabotropic glutamate receptors (mGluRs) suppressing neurotransmitter release in rodents, but their effects in human neocortex are unknown. We tested whether activation of group III mGluRs by L-AP4 changes GABAA receptor-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) in 2 distinct dendritic spine-innervating GABAergic interneurons recorded in vitro in human neocortex. Calbindin-positive double bouquet cells (DBCs) had columnar "horsetail" axons descending through layers II-V innervating dendritic spines (48%) and shafts, but not somata of pyramidal and nonpyramidal neurons. Parvalbumin-expressing dendrite-targeting cell (PV-DTC) axons extended in all directions innervating dendritic spines (22%), shafts (65%), and somata (13%). As measured, 20% of GABAergic neuropil synapses innervate spines, hence DBCs, but not PV-DTCs, preferentially select spine targets. Group III mGluR activation paradoxically increased the frequency of sIPSCs in DBCs (to median 137% of baseline) but suppressed it in PV-DTCs (median 92%), leaving the amplitude unchanged. The facilitation of sIPSCs in DBCs may result from their unique GABAergic input being disinhibited via network effect. We conclude that dendritic spines receive specialized, diverse GABAergic inputs, and group III mGluRs differentially regulate GABAergic synaptic transmission to distinct GABAergic cell types in human cortex.
Collapse
Affiliation(s)
- Istvan P Lukacs
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | | | - Martin Field
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Emily Hunter
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Michael Howarth
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Sawa Horie
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Puneet Plaha
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Richard Stacey
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Laurent Livermore
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Gabor Tamas
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, 6726 Szeged, Hungary
| | - Peter Somogyi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
5
|
Masugi-Tokita M, Kubota S, Kobayashi K, Yoshida T, Kageyama S, Sakamoto H, Kawauchi A. Spinal Transection Switches the Effect of Metabotropic Glutamate Receptor Subtype 7 from the Facilitation to Inhibition of Ejaculation. Neuroscience 2023; 509:10-19. [PMID: 36403690 DOI: 10.1016/j.neuroscience.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/03/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Metabotropic glutamate receptor subtype 7 (mGluR7) is a member of the group III mGluRs, which localize to presynaptic active zones of the central nervous system. We previously reported that mGluR7 knockout (KO) mice exhibit ejaculatory disorders, although they have normal sexual motivation. We hypothesized that mGluR7 regulates ejaculation by potentiating the excitability of the neural circuit in the lumbosacral spinal cord, because administration of the mGluR7-selective antagonist into that region inhibits drug-induced ejaculation. In the present study, to elucidate the mechanism of impaired ejaculation in mGluR7 KO mice, we eliminated the influence of the brain by spinal transection (spinalization). Unexpectedly, sexual responses of male mGluR7 KO mice were stronger than those of wild-type mice after spinalization. Histological examination indicated that mGluR7 controls sympathetic neurons as well as parasympathetic neurons. In view of the complexity of its synaptic regulation, mGluR7 might control ejaculation by multi-level and multi-modal mechanisms. Our study provides insight into the mechanism of ejaculation as well as a strategy for future therapies to treat ejaculatory disorders in humans.
Collapse
Affiliation(s)
- Miwako Masugi-Tokita
- Department of Urology, Shiga University of Medical Science, Otsu, Japan; World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan; Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| | - Shigehisa Kubota
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Kenichi Kobayashi
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Tetsuya Yoshida
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Susumu Kageyama
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Hirotaka Sakamoto
- Ushimado Marine Institute (UMI), Graduate School of Natural Science and Technology, Okayama University, Setouchi, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
6
|
The mGlu 7 receptor in schizophrenia - An update and future perspectives. Pharmacol Biochem Behav 2022; 218:173430. [PMID: 35870668 DOI: 10.1016/j.pbb.2022.173430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022]
Abstract
The mGlu7 receptor belongs to the III group of metabotropic glutamatergic (mGlu) receptors and physiologically serves as an "emergency" receptor that is activated by high, almost pathological, glutamate concentrations. Of all mGlu receptors, this receptor is most highly expressed in the brain. Additionally, relatively intense expression of the receptor was found at the periphery, for example in the bowels or in the reproductive system of male mice, but this review will be focused predominantly on its role in the brain. In the CNS, the receptor is expressed presynaptically, in the center of the synaptic cleft, at the terminals of both excitatory glutamatergic and inhibitory GABAergic neurons. Thus, it may regulate the release of both glutamate and GABA. Schizophrenia is thought to develop as a consequence of a disturbed glutamatergic-GABAergic balance in different parts of the brain. Thus, the mGlu7 receptor may be involved in the pathophysiology of schizophrenia and consequently constitute the target for antipsychotic drug discovery. In this review, we summarize the available data about mGlu7 receptor ligands and their activity in animal models of schizophrenia. At present, only a few ligands are available, and negative allosteric modulators (NAMs) appear to exert antipsychotic-like efficacy, indicating that the inhibition of the receptor could constitute a promising target in the search for novel drugs. Additionally, the data concerning the expression of the receptor in the CNS and putative mechanisms by which its inhibition may contribute to the treatment of schizophrenia will be discussed. Finally, the polymorphisms of genes encoding the receptor in schizophrenic patients will also be provided.
Collapse
|
7
|
Membrane trafficking and positioning of mGluRs at presynaptic and postsynaptic sites of excitatory synapses. Neuropharmacology 2021; 200:108799. [PMID: 34592242 DOI: 10.1016/j.neuropharm.2021.108799] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 01/21/2023]
Abstract
The plethora of functions of glutamate in the brain are mediated by the complementary actions of ionotropic and metabotropic glutamate receptors (mGluRs). The ionotropic glutamate receptors carry most of the fast excitatory transmission, while mGluRs modulate transmission on longer timescales by triggering multiple intracellular signaling pathways. As such, mGluRs mediate critical aspects of synaptic transmission and plasticity. Interestingly, at synapses, mGluRs operate at both sides of the cleft, and thus bidirectionally exert the effects of glutamate. At postsynaptic sites, group I mGluRs act to modulate excitability and plasticity. At presynaptic sites, group II and III mGluRs act as auto-receptors, modulating release properties in an activity-dependent manner. Thus, synaptic mGluRs are essential signal integrators that functionally couple presynaptic and postsynaptic mechanisms of transmission and plasticity. Understanding how these receptors reach the membrane and are positioned relative to the presynaptic glutamate release site are therefore important aspects of synapse biology. In this review, we will discuss the currently known mechanisms underlying the trafficking and positioning of mGluRs at and around synapses, and how these mechanisms contribute to synaptic functioning. We will highlight outstanding questions and present an outlook on how recent technological developments will move this exciting research field forward.
Collapse
|
8
|
McCullock TW, Kammermeier PJ. The evidence for and consequences of metabotropic glutamate receptor heterodimerization. Neuropharmacology 2021; 199:108801. [PMID: 34547332 DOI: 10.1016/j.neuropharm.2021.108801] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022]
Abstract
Metabotropic glutamate receptors (mGluRs) are an essential component of the mammalian central nervous system. These receptors modulate neuronal excitability in response to extracellular glutamate through the activation of intracellular heterotrimeric G proteins. Like most other class C G protein-coupled receptors, mGluRs function as obligate dimer proteins, meaning they need to form dimer complexes before becoming functional receptors. All mGluRs possess the ability to homodimerize, but studies over the past ten years have demonstrated these receptors are also capable of forming heterodimers in specific patterns. These mGluR heterodimers appear to have their own unique biophysical behavior and pharmacology with both native and synthetic compounds with few rules having been identified that allow for prediction of the consequences of any particular mGluR pair forming heterodimers. Here, we review the relevant literature demonstrating the existence and consequences of mGluR heterodimerization. By collecting biophysical and pharmacological data of several mGluR heterodimers we demonstrate the lack of generalizable behavior of these complexes indicating that each individual dimeric pair needs to be investigated independently. Additionally, by combining sequence alignment and structural analysis, we propose that interactions between the β4-A Helix Loop and the D Helix in the extracellular domain of these receptors are the structural components that dictate heterodimerization compatibility. Finally, we discuss the potential implications of mGluR heterodimerization from the viewpoints of further developing our understanding of neuronal physiology and leveraging mGluRs as a therapeutic target for the treatment of pathophysiology.
Collapse
Affiliation(s)
- Tyler W McCullock
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA.
| | - Paul J Kammermeier
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA.
| |
Collapse
|
9
|
Targeting metabotropic glutamate receptors for the treatment of depression and other stress-related disorders. Neuropharmacology 2021; 196:108687. [PMID: 34175327 DOI: 10.1016/j.neuropharm.2021.108687] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022]
Abstract
The discovery of robust antidepressant effects of ketamine in refractory patients has led to increasing focus on agents targeting glutamatergic signaling as potential novel antidepressant strategy. Among the agents targeting the glutamatergic system, compounds acting at metabotropic glutamate (mGlu) receptors are among the most promising agents under studies for depressive disorders. Further, the receptor diversity, distinct distribution in the CNS, and ability to modulate the glutamatergic neurotransmission in the brain areas implicated in mood disorders make them an exciting target for stress-related disorders. In preclinical models, antidepressant and anxiolytic effects of mGlu5 negative allosteric modulators (NAMs) have been reported. Interestingly, mGlu2/3 receptor antagonists show fast and sustained antidepressant-like effects similar to that of ketamine in rodents. Excitingly, they can also induce antidepressant effects in the animal models of treatment-resistant depression and are devoid of the side-effects associated with ketamine. Unfortunately, clinical trials of both mGlu5 and mGlu2/3 receptor NAMs have been inconclusive, and additional trials using other compounds with suitable preclinical and clinical properties are needed. Although group III mGlu receptors have gained less attention, mGlu7 receptor ligands have been shown to induce antidepressant-like effects in rodents. Collectively, compounds targeting mGlu receptors provide an alternative approach to fill the outstanding clinical need for safer and more efficacious antidepressants. This article is part of the special Issue on "Glutamate Receptors - mGluRs".
Collapse
|
10
|
Olivero G, Vergassola M, Cisani F, Roggeri A, Pittaluga A. Presynaptic Release-regulating Metabotropic Glutamate Receptors: An Update. Curr Neuropharmacol 2021; 18:655-672. [PMID: 31775600 PMCID: PMC7457419 DOI: 10.2174/1570159x17666191127112339] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/15/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors represent the largest family of glutamate receptors in mammals and act as fine tuners of the chemical transmission in central nervous system (CNS). In the last decade, results concerning the expression and the subcellular localization of mGlu receptors further clarified their role in physio-pathological conditions. Concomitantly, their pharmacological characterization largely improved thanks to the identification of new compounds (chemical ligands and antibodies recognizing epitopic sequences of the receptor proteins) that allowed to decipher the protein compositions of the naive receptors. mGlu receptors are expressed at the presynaptic site of chemical synapses. Here, they modulate intraterminal enzymatic pathways controlling the migration and the fusion of vesicles to synaptic membranes as well as the phosphorylation of colocalized receptors. Both the control of transmitter exocytosis and the phosphorylation of colocalized receptors elicited by mGlu receptors are relevant events that dictate the plasticity of nerve terminals, and account for the main role of presynaptic mGlu receptors as modulators of neuronal signalling. The role of the presynaptic mGlu receptors in the CNS has been the matter of several studies and this review aims at briefly summarizing the recent observations obtained with isolated nerve endings (we refer to as synaptosomes). We focus on the pharmacological characterization of these receptors and on their receptor-receptor interaction / oligo-dimerization in nerve endings that could be relevant to the development of new therapeutic approaches for the cure of central pathologies.
Collapse
Affiliation(s)
| | | | | | | | - Anna Pittaluga
- Department of Pharmacy, University of Genoa, Genoa, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
11
|
Kryszkowski W, Boczek T. The G Protein-Coupled Glutamate Receptors as Novel Molecular Targets in Schizophrenia Treatment-A Narrative Review. J Clin Med 2021; 10:jcm10071475. [PMID: 33918323 PMCID: PMC8038150 DOI: 10.3390/jcm10071475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 12/02/2022] Open
Abstract
Schizophrenia is a severe neuropsychiatric disease with an unknown etiology. The research into the neurobiology of this disease led to several models aimed at explaining the link between perturbations in brain function and the manifestation of psychotic symptoms. The glutamatergic hypothesis postulates that disrupted glutamate neurotransmission may mediate cognitive and psychosocial impairments by affecting the connections between the cortex and the thalamus. In this regard, the greatest attention has been given to ionotropic NMDA receptor hypofunction. However, converging data indicates metabotropic glutamate receptors as crucial for cognitive and psychomotor function. The distribution of these receptors in the brain regions related to schizophrenia and their regulatory role in glutamate release make them promising molecular targets for novel antipsychotics. This article reviews the progress in the research on the role of metabotropic glutamate receptors in schizophrenia etiopathology.
Collapse
Affiliation(s)
- Waldemar Kryszkowski
- General Psychiatric Ward, Babinski Memorial Hospital in Lodz, 91229 Lodz, Poland;
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, 92215 Lodz, Poland
- Correspondence:
| |
Collapse
|
12
|
Cisani F, Olivero G, Usai C, Van Camp G, Maccari S, Morley-Fletcher S, Pittaluga AM. Antibodies Against the NH 2-Terminus of the GluA Subunits Affect the AMPA-Evoked Releasing Activity: The Role of Complement. Front Immunol 2021; 12:586521. [PMID: 33717067 PMCID: PMC7952438 DOI: 10.3389/fimmu.2021.586521] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/15/2021] [Indexed: 01/31/2023] Open
Abstract
Antibodies recognizing the amino-terminal domain of receptor subunit proteins modify the receptor efficiency to controlling transmitter release in isolated nerve endings (e.g., synaptosomes) indirectly confirming their presence in these particles but also allowing to speculate on their subunit composition. Western blot analysis and confocal microscopy unveiled the presence of the GluA1, GluA2, GluA3, and GluA4 receptor subunits in cortical synaptosomes. Functional studies confirmed the presence of presynaptic release-regulating AMPA autoreceptors in these terminals, whose activation releases [3H]D-aspartate ([3H]D-Asp, here used as a marker of glutamate) in a NBQX-dependent manner. The AMPA autoreceptors traffic in a constitutive manner, since entrapping synaptosomes with the pep2-SVKI peptide (which interferes with the GluA2-GRIP1/PICK1 interaction) amplified the AMPA-evoked releasing activity, while the inactive pep2-SVKE peptide was devoid of activity. Incubation of synaptosomes with antibodies recognizing the NH2 terminus of the GluA2 and the GluA3 subunits increased, although to a different extent, the GluA2 and 3 densities in synaptosomal membranes, also amplifying the AMPA-evoked glutamate release in a NBQX-dependent fashion. We then analyzed the releasing activity of complement (1:300) from both treated and untreated synaptosomes and found that the complement-induced overflow occurred in a DL-t-BOA-sensitive, NBQX-insensitive fashion. We hypothesized that anti-GluA/GluA complexes in neuronal membranes could trigger the classic pathway of activation of the complement, modifying its releasing activity. Accordingly, the complement-evoked release of [3H]D-Asp from antiGluA2 and anti-GluA3 antibody treated synaptosomes was significantly increased when compared to untreated terminals and facilitation was prevented by omitting the C1q component of the immunocomplex. Antibodies recognizing the NH2 terminus of the GluA1 or the GluA4 subunits failed to affect both the AMPA and the complement-evoked tritium overflow. Our results suggest the presence of GluA2/GluA3-containing release-regulating AMPA autoreceptors in cortical synaptosomes. Incubation of synaptosomes with commercial anti-GluA2 or anti-GluA3 antibodies amplifies the AMPA-evoked exocytosis of glutamate through a complement-independent pathway, involving an excessive insertion of AMPA autoreceptors in plasma membranes but also affects the complement-dependent releasing activity, by promoting the classic pathway of activation of the immunocomplex. Both events could be relevant to the development of autoimmune diseases typified by an overproduction of anti-GluA subunits.
Collapse
Affiliation(s)
- Francesca Cisani
- Pharmacology and Toxicology Section, Department of Pharmacy, DIFAR, Genoa, Italy
| | - Guendalina Olivero
- Pharmacology and Toxicology Section, Department of Pharmacy, DIFAR, Genoa, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council, Genoa, Italy
| | - Gilles Van Camp
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
- International Associated Laboratory (LIA), “Prenatal Stress and Neurodegenerative Diseases”, University of Lille – CNRS, UGSF UMR 8576/Sapienza University of Rome and IRCCS Neuromed, Lille, France
| | - Stefania Maccari
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
- International Associated Laboratory (LIA), “Prenatal Stress and Neurodegenerative Diseases”, University of Lille – CNRS, UGSF UMR 8576/Sapienza University of Rome and IRCCS Neuromed, Lille, France
- Department of Science and Medical - Surgical Biotechnology, University Sapienza of Rome, Rome, Italy
| | - Sara Morley-Fletcher
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
- International Associated Laboratory (LIA), “Prenatal Stress and Neurodegenerative Diseases”, University of Lille – CNRS, UGSF UMR 8576/Sapienza University of Rome and IRCCS Neuromed, Lille, France
| | - Anna Maria Pittaluga
- Pharmacology and Toxicology Section, Department of Pharmacy, DIFAR, Genoa, Italy
- IRCCS San Martino Hospital, Genova, Italy
| |
Collapse
|
13
|
Fisher NM, AlHashim A, Buch AB, Badivuku H, Samman MM, Weiss KM, Cestero GI, Does MD, Rook JM, Lindsley CW, Conn PJ, Gogliotti RG, Niswender CM. A GRM7 mutation associated with developmental delay reduces mGlu7 expression and produces neurological phenotypes. JCI Insight 2021; 6:143324. [PMID: 33476302 PMCID: PMC7934925 DOI: 10.1172/jci.insight.143324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/13/2021] [Indexed: 12/29/2022] Open
Abstract
The metabotropic glutamate receptor 7 (mGlu7) is a G protein–coupled receptor that has been recently linked to neurodevelopmental disorders. This association is supported by the identification of GRM7 variants in patients with autism spectrum disorder, attention deficit hyperactivity disorder, and severe developmental delay. One GRM7 mutation previously reported in 2 patients results in a single amino acid change, I154T, within the mGlu7 ligand-binding domain. Here, we report 2 new patients with this mutation who present with severe developmental delay and epilepsy. Functional studies of the mGlu7-I154T mutant reveal that this substitution resulted in significant loss of mGlu7 protein expression in HEK293A cells and in mice. We show that this occurred posttranscriptionally at the level of protein expression and trafficking. Similar to mGlu7–global KO mice, mGlu7-I154T animals exhibited reduced motor coordination, deficits in contextual fear learning, and seizures. This provides functional evidence that a disease-associated mutation affecting the mGlu7 receptor was sufficient to cause neurological dysfunction in mice and further validates GRM7 as a disease-causing gene in the human population.
Collapse
Affiliation(s)
- Nicole M Fisher
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Aditi B Buch
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Hana Badivuku
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Kelly M Weiss
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Gabriela I Cestero
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Mark D Does
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jerri M Rook
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Craig W Lindsley
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Department of Chemistry and.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - P Jeffrey Conn
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee USA
| | - Rocco G Gogliotti
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, Illinois, USA
| | - Colleen M Niswender
- Department of Pharmacology and.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee USA
| |
Collapse
|
14
|
Cisani F, Roggeri A, Olivero G, Garrone B, Tongiani S, Di Giorgio FP, Pittaluga A. Acute Low Dose of Trazodone Recovers Glutamate Release Efficiency and mGlu2/3 Autoreceptor Impairments in the Spinal Cord of Rats Suffering From Chronic Sciatic Ligation. Front Pharmacol 2020; 11:1108. [PMID: 32765286 PMCID: PMC7379891 DOI: 10.3389/fphar.2020.01108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/08/2020] [Indexed: 11/16/2022] Open
Abstract
We investigated whether chronic sciatic ligation modifies the glutamate release in spinal cord nerve endings (synaptosomes) as well as the expression and the function of presynaptic release-regulating mGlu2/3 autoreceptors and 5-HT2A heteroreceptors in these particles. Synaptosomes were from the spinal cord of animals suffering from the sciatic ligation that developed on day 6 post-surgery a significant decrease of the force inducing paw-withdrawal in the lesioned paw. The exocytosis of glutamate (quantified as release of preloaded [3H]D-aspartate, [3H]D-Asp) elicited by a mild depolarizing stimulus (15 mM KCl) was significantly increased in synaptosomes from injured rats when compared to controls (uninjured rats). The mGlu2/3 agonist LY379268 (1000 pM) significantly inhibited the 15 mM KCl-evoked [3H]D-Asp overflow from control synaptosomes, but not in terminals isolated from injured animals. Differently, a low concentration (10 nM) of (±) DOI, unable to modify the 15 mM KCl-evoked [3H]D-Asp overflow in control spinal cord synaptosomes, significantly reduced the glutamate exocytosis in nerve endings isolated from the injured rats. Acute oral trazodone (TZD, 0.3 mg/kg on day 7 post-surgery) efficiently recovered glutamate exocytosis as well as the efficiency of LY379268 in inhibiting this event in spinal cord synaptosomes from injured animals. The sciatic ligation significantly reduced the expression of mGlu2/3, but not of 5-HT2A, receptor proteins in spinal cord synaptosomal lysates. Acute TZD recovered this parameter. Our results support the use of 5-HT2A antagonists for restoring altered spinal cord glutamate plasticity in rats suffering from sciatic ligation.
Collapse
Affiliation(s)
- Francesca Cisani
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Alessandra Roggeri
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Guendalina Olivero
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Beatrice Garrone
- Angelini RR&D (Research, Regulatory & Development), Angelini Pharma S.p.A., Rome, Italy
| | - Serena Tongiani
- Angelini RR&D (Research, Regulatory & Development), Angelini Pharma S.p.A., Rome, Italy
| | | | - Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
15
|
Shortall SE, Brown AM, Newton-Mann E, Dawe-Lane E, Evans C, Fowler M, King MV. Calbindin Deficits May Underlie Dissociable Effects of 5-HT 6 and mGlu 7 Antagonists on Glutamate and Cognition in a Dual-Hit Neurodevelopmental Model for Schizophrenia. Mol Neurobiol 2020; 57:3439-3457. [PMID: 32533466 PMCID: PMC7340678 DOI: 10.1007/s12035-020-01938-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022]
Abstract
Despite several compounds entering clinical trials for the negative and cognitive symptoms of schizophrenia, few have progressed beyond phase III. This is partly attributed to a need for improved preclinical models, to understand disease and enable predictive evaluation of novel therapeutics. To this end, one recent approach incorporates "dual-hit" neurodevelopmental insults like neonatal phencyclidine plus isolation rearing (PCP-Iso). Glutamatergic dysfunction contributes to schizophrenia pathophysiology and may represent a treatment target, so we used enzyme-based microsensors to evaluate basal- and drug-evoked glutamate release in hippocampal slices from rats that received neonatal PCP and/or isolation rearing. 5-HT6 antagonist-evoked glutamate release (thought to be mediated indirectly via GABAergic disinhibition) was reduced in PCP-Iso, as were cognitive effects of a 5-HT6 antagonist in a hippocampal glutamate-dependent novel object discrimination task. Yet mGlu7 antagonist-evoked glutamatergic and cognitive responses were spared. Immunohistochemical analyses suggest these findings (which mirror the apparent lack of clinical response to 5-HT6 antagonists in schizophrenia) are not due to reduced hippocampal 5-HT input in PCP-Iso, but may be explained by reduced calbindin expression. This calcium-binding protein is present in a subset of GABAergic interneurons receiving preferential 5-HT innervation and expressing 5-HT6 receptors. Its loss (in schizophrenia and PCP-Iso) would be expected to reduce interneuron firing and potentially prevent further 5-HT6 antagonist-mediated disinhibition, without impacting on responses of VIP-expressing interneurons to mGlu7 antagonism. This research highlights the importance of improved understanding for selection of appropriate preclinical models, especially where disease neurobiology impacts on cells mediating the effects of potential therapeutics.
Collapse
Affiliation(s)
- Sinead E Shortall
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Angus M Brown
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Eliot Newton-Mann
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Erin Dawe-Lane
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Chanelle Evans
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Maxine Fowler
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Madeleine V King
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
16
|
Fisher NM, Gould RW, Gogliotti RG, McDonald AJ, Badivuku H, Chennareddy S, Buch AB, Moore AM, Jenkins MT, Robb WH, Lindsley CW, Jones CK, Conn PJ, Niswender CM. Phenotypic profiling of mGlu 7 knockout mice reveals new implications for neurodevelopmental disorders. GENES BRAIN AND BEHAVIOR 2020; 19:e12654. [PMID: 32248644 DOI: 10.1111/gbb.12654] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/03/2020] [Accepted: 03/26/2020] [Indexed: 12/17/2022]
Abstract
Neurodevelopmental disorders are characterized by deficits in communication, cognition, attention, social behavior and/or motor control. Previous studies have pointed to the involvement of genes that regulate synaptic structure and function in the pathogenesis of these disorders. One such gene, GRM7, encodes the metabotropic glutamate receptor 7 (mGlu7 ), a G protein-coupled receptor that regulates presynaptic neurotransmitter release. Mutations and polymorphisms in GRM7 have been associated with neurodevelopmental disorders in clinical populations; however, limited preclinical studies have evaluated mGlu7 in the context of this specific disease class. Here, we show that the absence of mGlu7 in mice is sufficient to alter phenotypes within the domains of social behavior, associative learning, motor function, epilepsy and sleep. Moreover, Grm7 knockout mice exhibit an attenuated response to amphetamine. These findings provide rationale for further investigation of mGlu7 as a potential therapeutic target for neurodevelopmental disorders such as idiopathic autism, attention deficit hyperactivity disorder and Rett syndrome.
Collapse
Affiliation(s)
- Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Robert W Gould
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Rocco G Gogliotti
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Annalise J McDonald
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Hana Badivuku
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Susmita Chennareddy
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Aditi B Buch
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Annah M Moore
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Matthew T Jenkins
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - W Hudson Robb
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Morales-Weil K, Moreno M, Ahumada J, Arriagada J, Fuentealba P, Bonansco C, Fuenzalida M. Priming of GABAergic Long-term Potentiation by Muscarinic Receptors. Neuroscience 2020; 428:242-251. [PMID: 31917346 DOI: 10.1016/j.neuroscience.2019.12.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 11/25/2019] [Accepted: 12/19/2019] [Indexed: 12/28/2022]
Abstract
Growing evidence indicates that GABAergic interneurons play a pivotal role to generate brain oscillation patterns, which are fundamental for the mnemonic processing of the hippocampus. While acetylcholine (ACh) is a powerful modulator of synaptic plasticity and brain function, few studies have been focused on the role of cholinergic signaling in the regulation of GABAergic inhibitory synaptic plasticity. We have previously shown that co-activation of endocannabinoids (CB1R) and muscarinic receptor (mAChR) in hippocampal interneurons can induce activity-dependent GABAergic long-term depression in CA1 pyramidal neurons. Here, using electrophysiological and pharmacological approaches in acute rat hippocampal slices, we show that activation of cholinergic receptors followed by either high-frequency stimulation of Schaeffer collaterals or exogenous activation of metabotropic glutamate receptor (mGluR) induces a robust long-term potentiation at GABAergic synapses (iLTP). These forms of iLTP are blocked by the M1 type of mAChR (MR1) or by the group I of mGluR (mGluR1/5) antagonists. These results suggest the existence of spatiotemporal cooperativity between cholinergic and glutamatergic pathways where activation of mAChR serves as a metaplastic switch making glutamatergic synapses capable to induce long-term potentiation at inhibitory synapses, that may contribute to the modulation of brain mechanisms of learning and memory.
Collapse
Affiliation(s)
- Koyam Morales-Weil
- Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Chile; Programa de Doctorado en Ciencias, mención Neurociencias, Universidad de Valparaíso, Chile
| | - Macarena Moreno
- Centro interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Chile
| | - Juan Ahumada
- Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Chile; Programa de Doctorado en Ciencias, mención Neurociencias, Universidad de Valparaíso, Chile
| | - Jorge Arriagada
- Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Chile
| | - Pablo Fuentealba
- Departamento de Psiquiatría, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Chile
| | - Christian Bonansco
- Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Chile.
| |
Collapse
|
18
|
Kasatkina LA, Tarasenko AS, Krupko OO, Kuchmerovska TM, Lisakovska OO, Trikash IO. Vitamin D deficiency induces the excitation/inhibition brain imbalance and the proinflammatory shift. Int J Biochem Cell Biol 2019; 119:105665. [PMID: 31821883 DOI: 10.1016/j.biocel.2019.105665] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/25/2019] [Accepted: 12/05/2019] [Indexed: 12/16/2022]
Abstract
Vitamin D3 is among the major neurosteroids whose role in developing and adult brain is intensively studied now. Its active form 1,25(OH)2D3 regulates the expression and functioning of a range of brain-specific proteins, which orchestrate the neurotransmitter turnover, neurogenesis and neuroplasticity. Despite numerous studies of the vitamin D role in normal and pathological brain function, there is little evidence on the mechanisms of alterations in excitatory and inhibitory neurotransmission under vitamin D deficiency (VDD). Using the animal model we characterized the dysfunction of excitatory and inhibitory neurotransmission under alimentary VDD. The shift between unstimulated and evoked GABA release under VDD was largely reversed after treatment of VDD, whereas the impairments in glutamatergic system were only partially recovered after 1-month vitamin D3 supplementation. The increase of the external glutamate level and unstimulated GABA release in brain nerve terminals was associated with intensified ROS production and higher [Ca2+]i in presynapse. The negative allosteric modulation of presynaptic mGlu7 receptors significantly enhanced exocytotic GABA release, which was decreased under VDD, thereby suggesting the neuroprotective effect of such modulation of inhibitory neurotransmission. Synaptic plasma membranes and cytosolic proteins contribute to the decreased stimulated release of neurotransmitter, by being the crucial components, whose functional state is impaired under VDD. The critical changes with synaptic vesicles occurred at the docking step of the process, whereas malfunctioning of synaptic cytosolic proteins impacted the fusion event foremost. The decreased amplitude of exocytosis was inherent for non-excitable cells as well, as evidenced by lower platelet degranulation. Our data suggest the presynaptic dysfunction and proinflammatory shift as the early events in the pathogenesis of VDD-associated disorders and provide evidences for the neuroprotective role of vitamin D3.
Collapse
Affiliation(s)
- Ludmila A Kasatkina
- The Department of Neurochemistry, Palladin Institute of Biochemistry, NAS of Ukraine, 9, Leontovycha Street, Kyiv, 01030, Ukraine
| | - Alla S Tarasenko
- The Department of Neurochemistry, Palladin Institute of Biochemistry, NAS of Ukraine, 9, Leontovycha Street, Kyiv, 01030, Ukraine
| | - Olga O Krupko
- The Department of Neurochemistry, Palladin Institute of Biochemistry, NAS of Ukraine, 9, Leontovycha Street, Kyiv, 01030, Ukraine
| | - Tamara M Kuchmerovska
- The Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, NAS of Ukraine, 9, Leontovycha Street, Kyiv, 01030 Ukraine
| | - Olha O Lisakovska
- The Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, NAS of Ukraine, 9, Leontovycha Street, Kyiv, 01030 Ukraine
| | - Irene O Trikash
- The Department of Neurochemistry, Palladin Institute of Biochemistry, NAS of Ukraine, 9, Leontovycha Street, Kyiv, 01030, Ukraine.
| |
Collapse
|
19
|
Pittaluga A. Acute Functional Adaptations in Isolated Presynaptic Terminals Unveil Synaptosomal Learning and Memory. Int J Mol Sci 2019; 20:ijms20153641. [PMID: 31349638 PMCID: PMC6696074 DOI: 10.3390/ijms20153641] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/08/2019] [Accepted: 07/24/2019] [Indexed: 01/19/2023] Open
Abstract
Synaptosomes are used to decipher the mechanisms involved in chemical transmission, since they permit highlighting the mechanisms of transmitter release and confirming whether the activation of presynaptic receptors/enzymes can modulate this event. In the last two decades, important progress in the field came from the observations that synaptosomes retain changes elicited by both “in vivo” and “in vitro” acute chemical stimulation. The novelty of these studies is the finding that these adaptations persist beyond the washout of the triggering drug, emerging subsequently as functional modifications of synaptosomal performances, including release efficiency. These findings support the conclusion that synaptosomes are plastic entities that respond dynamically to ambient stimulation, but also that they “learn and memorize” the functional adaptation triggered by acute exposure to chemical agents. This work aims at reviewing the results so far available concerning this form of synaptosomal learning, also highlighting the role of these acute chemical adaptations in pathological conditions.
Collapse
Affiliation(s)
- Anna Pittaluga
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, Viale Cembrano 4, 16148 and Center of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV, 16132 University of Genoa, 16145 Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, 16145, Genova, Italy.
| |
Collapse
|
20
|
Impact of the metabotropic glutamate receptor7 (mGlu 7) allosteric agonist, AMN082, on fear learning and memory and anxiety-like behavior. Eur J Pharmacol 2019; 858:172512. [PMID: 31260653 DOI: 10.1016/j.ejphar.2019.172512] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/19/2019] [Accepted: 06/27/2019] [Indexed: 12/30/2022]
Abstract
The present study was conducted to evaluate the influence of AMN082, the metabotropic glutamate receptor subtype 7 (mGlu7) allosteric agonist on different stages of memory processes connected with fear conditioning in the passive avoidance (PA) learning task in mice and negative emotional state (anxiety-like) induced by ethanol- and morphine-withdrawal in the elevated plus maze (EPM) test in rats. To perform the PA test, AMN082 (1.25, 2.5 and 5 mg/kg, i. p.) was injected to interfere with (or inhibit) acquisition, consolidation, and retrieval processes. The retention latency in each group was recorded using a step-through passive avoidance task 24 h after training. In turn, in ethanol- and morphine-withdrawal rats, the influence of AMN082 on anxiety-like behavior was estimated in the EPM test 24 h- (ethanol) and 72- h (morphine) after the last dose of repeated drug administrations. In all experimental groups, AMN082 at the dose of 5 mg/kg significantly decreased the step-through latency of long-term memory in the PA task. These AMN082 effects were reversed by MMPIP (10 mg/kg), the antagonist of mGlu7 receptor. AMN082 (2.5 and 5 mg/kg) also decreased ethanol- and morphine withdrawal-induced anxiety-like behavior in the EPM test, and this AMN082 (5 mg/kg) effect was counteracted by MMPIP pretreatment. Taken together, the results show that mGlu7 is involved in fear learning to the context and anxiety-like state connected with unpleasant experiences after ethanol- and morphine withdrawal in rodents. However, it appears that functional dissociation exists between these two AMN082 effects.
Collapse
|
21
|
Stachniak TJ, Sylwestrak EL, Scheiffele P, Hall BJ, Ghosh A. Elfn1-Induced Constitutive Activation of mGluR7 Determines Frequency-Dependent Recruitment of Somatostatin Interneurons. J Neurosci 2019; 39:4461-4474. [PMID: 30940718 PMCID: PMC6554623 DOI: 10.1523/jneurosci.2276-18.2019] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 02/14/2019] [Accepted: 03/22/2019] [Indexed: 11/21/2022] Open
Abstract
Excitatory synapses onto somatostatin (SOM) interneurons show robust short-term facilitation. This hallmark feature of SOM interneurons arises from a low initial release probability that regulates the recruitment of interneurons in response to trains of action potentials. Previous work has shown that Elfn1 (extracellular leucine rich repeat and fibronectin Type III domain containing 1) is necessary to generate facilitating synapses onto SOM neurons by recruitment of two separate presynaptic components: mGluR7 (metabotropic glutamate receptor 7) and GluK2-KARs (kainate receptors containing glutamate receptor, ionotropic, kainate 2). Here, we identify how a transsynaptic interaction between Elfn1 and mGluR7 constitutively reduces initial release probability onto mouse cortical SOM neurons. Elfn1 produces glutamate-independent activation of mGluR7 via presynaptic clustering, resulting in a divergence from the canonical "autoreceptor" role of Type III mGluRs, and substantially altering synaptic pharmacology. This structurally induced determination of initial release probability is present at both layer 2/3 and layer 5 synapses. In layer 2/3 SOM neurons, synaptic facilitation in response to spike trains is also dependent on presynaptic GluK2-KARs. In contrast, layer 5 SOM neurons do not exhibit presynaptic GluK2-KAR activity at baseline and show reduced facilitation. GluK2-KAR engagement at synapses onto layer 5 SOM neurons can be induced by calmodulin activation, suggesting that synaptic function can be dynamically regulated. Thus, synaptic facilitation onto SOM interneurons is mediated both by constitutive mGluR7 recruitment by Elfn1 and regulated GluK2-KAR recruitment, which determines the extent of interneuron recruitment in different cortical layers.SIGNIFICANCE STATEMENT This study identifies a novel mechanism for generating constitutive GPCR activity through a transsynaptic Elfn1/mGluR7 structural interaction. The resulting tonic suppression of synaptic release probability deviates from canonical autoreceptor function. Constitutive suppression delays the activation of somatostatin interneurons in circuits, necessitating high-frequency activity for somatostatin interneuron recruitment. Furthermore, variations in the synaptic proteome generate layer-specific differences in facilitation at pyr → SOM synapses. The presence of GluK2 kainate receptors in L2/3 enhances synaptic transmission during prolonged activity. Thus, layer-specific synaptic properties onto somatostatin interneurons are mediated by both constitutive mGluR7 recruitment and regulated GluK2 kainate receptor recruitment, revealing a mechanism that generates diversity in physiological responses of interneurons.
Collapse
Affiliation(s)
- Tevye Jason Stachniak
- F. Hoffmann-La Roche Ltd, Roche Innovation Center Basel, Basel 4051, Switzerland
- University of Basel, Departement Biozentrum, Basel 4056, Switzerland, and
- Biogen, Cambridge, Massachusetts 02142
| | - Emily Lauren Sylwestrak
- F. Hoffmann-La Roche Ltd, Roche Innovation Center Basel, Basel 4051, Switzerland
- Stanford University, Department of Bioengineering, Stanford, California 94305
- University of Basel, Departement Biozentrum, Basel 4056, Switzerland, and
| | - Peter Scheiffele
- University of Basel, Departement Biozentrum, Basel 4056, Switzerland, and
| | - Benjamin J Hall
- F. Hoffmann-La Roche Ltd, Roche Innovation Center Basel, Basel 4051, Switzerland
| | - Anirvan Ghosh
- F. Hoffmann-La Roche Ltd, Roche Innovation Center Basel, Basel 4051, Switzerland,
- Biogen, Cambridge, Massachusetts 02142
| |
Collapse
|
22
|
Chaki S, Koike H, Fukumoto K. Targeting of Metabotropic Glutamate Receptors for the Development of Novel Antidepressants. CHRONIC STRESS 2019; 3:2470547019837712. [PMID: 32500107 PMCID: PMC7243201 DOI: 10.1177/2470547019837712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 02/21/2019] [Indexed: 12/22/2022]
Abstract
Since discovering that ketamine has robust antidepressant effects, the
glutamatergic system has been proposed as an attractive target for the
development of novel antidepressants. Among the glutamatergic system,
metabotropic glutamate (mGlu) receptors are of interest because mGlu receptors
play modulatory roles in glutamatergic transmission, consequently, agents acting
on mGlu receptors might not exert the adverse effects associated with ketamine.
mGlu receptors have eight subtypes that are classified into three groups, and
the roles of each mGlu receptor subtype in depression are being investigated. To
date, the potential use of mGlu5 receptor antagonists and mGlu2/3 receptor
antagonists as antidepressants has been actively investigated, and the
mechanisms underlying these antidepressant effects are being delineated.
Although the outcomes of clinical trials using an mGlu5 receptor negative
allosteric modulator and an mGlu2/3 receptor negative allosteric modulator have
not been encouraging, these trials have been inconclusive, and additional trials
using other compounds with more appropriate profiles are needed. In contrast,
the roles of group III mGlu receptors have not yet been fully elucidated because
of a lack of suitable pharmacological tools. Nonetheless, investigations of the
use of mGlu4 and mGlu7 receptors as drug targets for the development of
antidepressants have been ongoing, and some interesting evidence has been
obtained.
Collapse
|
23
|
Olivero G, Vergassola M, Cisani F, Usai C, Pittaluga A. Immuno-Pharmacological Characterization of Presynaptic GluN3A-Containing NMDA Autoreceptors: Relevance to Anti-NMDA Receptor Autoimmune Diseases. Mol Neurobiol 2019; 56:6142-6155. [PMID: 30734226 DOI: 10.1007/s12035-019-1511-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/24/2019] [Indexed: 01/22/2023]
Abstract
Mouse hippocampal glutamatergic nerve endings express presynaptic release-regulating NMDA autoreceptors (NMDARs). The presence of GluN1, GluN2A, GluN2B, and GluN3A subunits in hippocampal vesicular glutamate transporter type 1-positive synaptosomes was confirmed with confocal microscopy. GluN2C, GluN2D, and GluN3B immunopositivity was scarcely present. Incubation of synaptosomes with the anti-GluN1, the anti-GluN2A, the anti-GluN2B, or the anti-GluN3A antibody prevented the 30 μM NMDA/1 μM glycine-evoked [3H]D-aspartate ([3H]D-ASP) release. The NMDA/glycine-evoked [3H]D-ASP release was reduced by increasing the external protons, consistent with the participation of GluN1 subunits lacking the N1 cassette to the receptor assembly. The result also excludes the involvement of GluN1/GluN3A dimers into the NMDA-evoked overflow. Complement (1:300) released [3H]D-ASP in a dizocilpine-sensitive manner, suggesting the participation of a NMDAR-mediated component in the releasing activity. Accordingly, the complement-evoked glutamate overflow was reduced in anti-GluN-treated synaptosomes when compared to the control. We speculated that incubation with antibodies had favored the internalization of NMDA receptors. Indeed, a significant reduction of the GluN1 and GluN2B proteins in the plasma membranes of anti-GluN1 or anti-GluN2B antibody-treated synaptosomes emerged in biotinylation studies. Altogether, our findings confirm the existence of presynaptic GluN3A-containing release-regulating NMDARs in mouse hippocampal glutamatergic nerve endings. Furthermore, they unveil presynaptic alteration of the GluN subunit insertion in synaptosomal plasma membranes elicited by anti-GluN antibodies that might be relevant to the central alterations occurring in patients suffering from autoimmune anti-NMDA diseases.
Collapse
Affiliation(s)
- Guendalina Olivero
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Matteo Vergassola
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Francesca Cisani
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council, via De Marini 6, 16149, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy. .,IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
24
|
O’Connor RM, McCafferty CP, Bravo JA, Singewald N, Holmes A, Cryan JF. Increased amygdalar metabotropic glutamate receptor 7 mRNA in a genetic mouse model of impaired fear extinction. Psychopharmacology (Berl) 2019; 236:265-272. [PMID: 30215216 PMCID: PMC6739849 DOI: 10.1007/s00213-018-5031-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/05/2018] [Indexed: 12/18/2022]
Abstract
RATIONALE Post-traumatic stress disorder (PTSD) is a devastating anxiety-related disorder which develops subsequent to a severe psychologically traumatic event. Only ~ 9% of people who experience such a trauma develop PTSD. It is clear that a number of factors, including genetics, influence whether an individual will develop PTSD subsequent to a trauma. The 129S1/SvImJ (S1) inbred mouse strain displays poor fear extinction and may be useful to model this specific aspect of PTSD. The metabotropic glutamate receptor 7 (mGlu7 receptor) has previously been shown to be involved in cognitive processes and anxiety-like behaviour placing it in a key position to regulate fear extinction processes. We sought to compare mGlu7 receptor mRNA levels in the S1 strain with those in the robustly extinguishing C57BL/6J (B6) inbred strain using in situ hybridisation (ISH) in three brain regions associated with fear extinction: the amygdala, hippocampus and prefrontal cortex (PFC). RESULTS Compared to the B6 strain, S1 mice had increased mGlu7 receptor mRNA levels in the lateral amygdala (LA) and basolateral amygdala (BLA) subdivisions. An increase was also seen in the hippocampal CA1 and CA3 subregions of S1 mice. No difference in mGlu7 receptor levels were seen in the central nucleus (CeA) of the amygdala, dentate gyrus (DG) of the hippocampus or prefrontal cortex. CONCLUSIONS These data show altered mGlu7 receptor expression in key brain regions associated with fear extinction in two different inbred mouse strains which differ markedly in their fear extinction behaviour. Altered mGlu7 receptor levels may contribute to the deficit fear extinction processes seen in fear extinction in the S1 strain.
Collapse
Affiliation(s)
- Richard M. O’Connor
- Department of Anatomy and Neuroscience and APC Microbiome Institute, University College Cork, Cork, Ireland,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, S10-20 Hess CSM, 1470 Madison Avenue, New York, NY 10029, USA,Present address: Department of Neuroscience, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Cian P. McCafferty
- Department of Anatomy and Neuroscience and APC Microbiome Institute, University College Cork, Cork, Ireland,Present address: Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Javier A. Bravo
- Grupo de NeuroGastroBioquímica, Laboratorio e Química Biológica & Bioquímica de Sistemas, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA
| | - John F. Cryan
- Department of Anatomy and Neuroscience and APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
25
|
Fisher NM, Seto M, Lindsley CW, Niswender CM. Metabotropic Glutamate Receptor 7: A New Therapeutic Target in Neurodevelopmental Disorders. Front Mol Neurosci 2018; 11:387. [PMID: 30405350 PMCID: PMC6206046 DOI: 10.3389/fnmol.2018.00387] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/01/2018] [Indexed: 12/27/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are characterized by a wide range of symptoms including delayed speech, intellectual disability, motor dysfunction, social deficits, breathing problems, structural abnormalities, and epilepsy. Unfortunately, current treatment strategies are limited and innovative new approaches are sorely needed to address these complex diseases. The metabotropic glutamate receptors are a class of G protein-coupled receptors that act to modulate neurotransmission across many brain structures. They have shown great promise as drug targets for numerous neurological and psychiatric diseases. Moreover, the development of subtype-selective allosteric modulators has allowed detailed studies of each receptor subtype. Here, we focus on the metabotropic glutamate receptor 7 (mGlu7) as a potential therapeutic target for NDDs. mGlu7 is expressed widely throughout the brain in regions that correspond to the symptom domains listed above and has established roles in synaptic physiology and behavior. Single nucleotide polymorphisms and mutations in the GRM7 gene have been associated with idiopathic autism and other NDDs in patients. In rodent models, existing literature suggests that decreased mGlu7 expression and/or function may lead to symptoms that overlap with those of NDDs. Furthermore, potentiation of mGlu7 activity has shown efficacy in a mouse model of Rett syndrome. In this review, we summarize current findings that provide rationale for the continued development of mGlu7 modulators as potential therapeutics.
Collapse
Affiliation(s)
- Nicole M Fisher
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Mabel Seto
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Craig W Lindsley
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.,Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Colleen M Niswender
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
26
|
Cieślik P, Woźniak M, Kaczorowska K, Brański P, Burnat G, Chocyk A, Bobula B, Gruca P, Litwa E, Pałucha-Poniewiera A, Wąsik A, Pilc A, Wierońska J. Negative Allosteric Modulators of mGlu 7 Receptor as Putative Antipsychotic Drugs. Front Mol Neurosci 2018; 11:316. [PMID: 30294258 PMCID: PMC6158327 DOI: 10.3389/fnmol.2018.00316] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/16/2018] [Indexed: 01/08/2023] Open
Abstract
The data concerning antipsychotic-like activity of negative allosteric modulators (NAMs)/antagonists of mGlu7 receptors are limited. The only available ligands for this receptor are MMPIP and ADX71743. In the present studies, we used stable cell line expressing mGlu7 receptor and it was shown that both compounds dose-dependently potentiated forskolin elevated cAMP concentration in the T-REx 293 cells, showing their inverse agonist properties. Subsequently, pharmacokinetic studies were performed. Both compounds were given intraperitoneally (i.p.) at the dose of 10 mg/kg and reached Cmax 0.25-0.5 h after administration, and then they declined rapidly, ADX71743 being almost undetectable 2 h after administration, while the concentration of MMPIP was still observed, suggesting that the concentration of MMPIP was more stable. Finally, we investigated the role of both mGlu7 receptor NAMs in animal models of schizophrenia. Behavioral tests commonly used in antipsychotic drug discovery were conducted. Both tested compounds dose-dependently inhibited MK-801-induced hyperactivity (MMPIP at 15 mg/kg; ADX at 5 and 15 mg/kg) and DOI-induced head twitches (MMPIP at 5, 10, 15 mg/kg; ADX at 2.5, 5, 10 mg/kg). Moreover, the same effects were noticed in novel object recognition test, where MMPIP (5, 10, 15 mg/kg) and ADX71743 (1, 5, 15 mg/kg) reversed MK-801-induced disturbances. In the social interaction test, antipsychotic activity was observed only for ADX71743 (5, 15 mg/kg). ADX71743 at the dose 2.5 mg/kg reversed MK-801-induced disruption in prepulse inhibition while MMPIP at 10 mg/kg reversed MK-801-induced disruption in spatial delayed alternation. The present studies showed that mGlu7 receptor may be considered as a putative target for antipsychotic drugs, though more studies are needed due to limited number of available ligands.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Joanna Wierońska
- Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
27
|
Vergassola M, Olivero G, Cisani F, Usai C, Bossi S, Puliti A, Pittaluga A. Presynaptic mGlu1 Receptors Control GABA B Receptors in an Antagonist-Like Manner in Mouse Cortical GABAergic and Glutamatergic Nerve Endings. Front Mol Neurosci 2018; 11:324. [PMID: 30279647 PMCID: PMC6153310 DOI: 10.3389/fnmol.2018.00324] [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: 07/06/2018] [Accepted: 08/22/2018] [Indexed: 01/03/2023] Open
Abstract
Mouse cortical GABAergic synaptosomes possess presynaptic inhibitory GABAB autoreceptors. Accordingly, (±)baclofen (3 μM) inhibits in a CGP53423-sensitive manner the 12 mM KCl-evoked release of preloaded [3H]GABA. Differently, the existence of presynaptic release-regulating metabotropic glutamate type 1 (mGlu1) heteroreceptors in these terminals is still matter of discussion, although confocal microscopy unveiled the existence of mGlu1α with GABAB1 or GABAB2 proteins in cortical VGAT-positive synaptosomes. The group I mGlu agonist 3,5-DHPG failed to modify on its own the 12 mM KCl-evoked [3H]GABA exocytosis from cortical nerve endings, but, when added concomitantly to the GABAB agonist, it significantly reduced the 3 μM (±)baclofen-induced inhibition of [3H]GABA exocytosis. Conversely, the mGlu1 antagonist LY367385 (0.03–1 μM), inactive on its own on GABA exocytosis, amplified the 3 μM (±)baclofen-induced inhibition of [3H]GABA overflow. The ( ± )baclofen-induced inhibition of [3H]GABA exocytosis was more pronounced in cortical synaptosomes from Grm1crv4/crv4 mice, which bear a spontaneous mutation of the Grm1 gene leading to the functional inactivation of the mGlu1 receptor. Inasmuch, the expression of GABAB2 receptor protein in cortical synaptosomal lysates from Grm1crv4/crv4 mice was increased when compared to controls. Altogether, these observations seem best interpreted by assuming that mGlu1 coexist with GABAB receptors in GABAergic cortical synaptosomes, where they control GABA receptors in an antagonist-like manner. We then asked whether the mGlu1-mediated control of GABAB receptors is restricted to GABAergic terminals, or if it occurs also in other subpopulations of nerve endings. Release-regulating GABAB receptors also exist in glutamatergic nerve endings. (±)baclofen (1 μM) diminished the 12 mM KCl-evoked [3H]D-aspartate overflow. Also in these terminals, the concomitant presence of 1 μM LY367385, inactive on its own, significantly amplified the inhibitory effect exerted by (±)baclofen on [3H]D-aspartate exocytosis. Confocal microscopy confirmed the colocalization of mGlu1 with GABAB1 and GABAB2 labeling in vesicular glutamate type1 transporter-positive particles. Our results support the conclusion that mGlu1 receptors modulate in an antagonist-like manner presynaptic release-regulating GABAB receptors. This receptor–receptor interaction could be neuroprotective in central disease typified by hyperglutamatergicity.
Collapse
Affiliation(s)
| | | | | | - Cesare Usai
- Institute of Biophysics, National Research Council, Genoa, Italy
| | - Simone Bossi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Aldamaria Puliti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.,IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
28
|
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
|
29
|
The reduction in glutamate release is predictive of cognitive and emotional alterations that are corrected by the positive modulator of AMPA receptors S 47445 in perinatal stressed rats. Neuropharmacology 2018; 135:284-296. [DOI: 10.1016/j.neuropharm.2018.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 12/22/2022]
|
30
|
Masugi-Tokita M, Yoshida T, Kageyama S, Kawata M, Kawauchi A. Metabotropic glutamate receptor subtype 7 has critical roles in regulation of the endocrine system and social behaviours. J Neuroendocrinol 2018; 30:e12575. [PMID: 29377390 DOI: 10.1111/jne.12575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 01/10/2018] [Accepted: 01/22/2018] [Indexed: 01/19/2023]
Abstract
Metabotropic glutamate receptor subtype 7 (mGluR7) is one of the group III mGluRs, which are negatively coupled to adenylate cyclase via Gi/Go proteins and localised to presynaptic active zones of the mammalian central nervous system. We previously reported that mGluR7 is essential for intermale aggression and amygdala-dependent fear learning. To elucidate the role of mGluR7 in the neuroendocrine system, we performed biochemical analyses and found a significant reduction of testosterone levels in mGluR7 knockout (KO) mice. Testosterone replacement restored intermale aggressive behaviour in castrated wild-type mice to the level of gonadally intact wild-type mice. However, given the same dosage of testosterone replacement, mGluR7 KO mice showed almost no aggressive behaviour. These results indicate that reduction of plasma testosterone is unrelated to the deficit in intermale aggression in mGluR7 KO mice. Social investigating behaviour of intact mGluR7 KO mice also differed from that of wild-type mice; e.g. the KO mice showing less frequent anogenital sniffing and more frequent grooming behaviour. Testosterone replacement increased anogenital sniffing and grooming behaviour in castrated mGluR7 KO mice, while the differences were still present between castrated wild-type mice and KO mice after both underwent testosterone replacement. These results imply that reduction of plasma testosterone may partially inhibit social investigating behaviours in intact mGluR7 KO mice. Furthermore, castrated mGluR7 KO mice have smaller seminal vesicles than those of castrated wild-type mice, although seminal vesicle weights were normal in intact mice. These observations suggest that, besides testicular testosterone, some other hormone levels may be dysregulated in mGluR7 KO mice, and indicate a critical role of mGluR7 in the endocrine system. Taken together, our findings demonstrate that mGluR7 is essential for the regulation of the endocrine system, in addition to innate behaviours such as intermale aggression and fear response.
Collapse
Affiliation(s)
- M Masugi-Tokita
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
- World Premier International Research Center Initiative-Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - T Yoshida
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - S Kageyama
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - M Kawata
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- School of Health Sciences, Bukkyo University, Kyoto, Japan
| | - A Kawauchi
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
31
|
Olivero G, Grilli M, Vergassola M, Bonfiglio T, Padolecchia C, Garrone B, Di Giorgio FP, Tongiani S, Usai C, Marchi M, Pittaluga A. 5-HT 2A-mGlu2/3 receptor complex in rat spinal cord glutamatergic nerve endings: A 5-HT 2A to mGlu2/3 signalling to amplify presynaptic mechanism of auto-control of glutamate exocytosis. Neuropharmacology 2018; 133:429-439. [PMID: 29499271 DOI: 10.1016/j.neuropharm.2018.02.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/23/2018] [Accepted: 02/26/2018] [Indexed: 11/25/2022]
Abstract
Presynaptic mGlu2/3 autoreceptors exist in rat spinal cord nerve terminals as suggested by the finding that LY379268 inhibited the 15 mM KCl-evoked release of [3H]D-aspartate ([3H]D-Asp) in a LY341495-sensitive manner. Spinal cord glutamatergic nerve terminals also possess presynaptic release-regulating 5-HT2A heteroreceptors. Actually, the 15 mM KCl-evoked [3H]D-Asp exocytosis from spinal cord synaptosomes was reduced by the 5-HT2A agonist (±)DOI, an effect reversed by the 5-HT2A antagonists MDL11,939, MDL100907, ketanserin and trazodone (TZD). We investigated whether mGlu2/3 and 5-HT2A receptors colocalize and cross-talk in these terminals and if 5-HT2A ligands modulate the mGlu2/3-mediated control of glutamate exocytosis. Western blot analysis and confocal microscopy highlighted the presence of mGlu2/3 and 5-HT2A receptor proteins in spinal cord VGLUT1 positive synaptosomes, where mGlu2/3 and 5-HT2A receptor immunoreactivities largely colocalize. Furthermore, mGlu2/3 immunoprecipitates from spinal cord synaptosomes were also 5-HT2A immunopositive. Interestingly, the 100 pM LY379268-induced reduction of the 15 mM KCl-evoked [3H]D-Asp overflow as well as its inhibition by 100 nM (±)DOI became undetectable when the two agonists were concomitantly added. Conversely, 5-HT2A antagonists (MDL11,939, MDL100907, ketanserin and TZD) reinforced the release-regulating activity of mGlu2/3 autoreceptors. Increased expression of mGlu2/3 receptor proteins in synaptosomal plasmamembranes paralleled the gain of function of the mGlu2/3 autoreceptors elicited by 5-HT2A antagonists. Based on these results, we propose that in spinal cord glutamatergic terminals i) mGlu2/3 and 5-HT2A receptors colocalize and interact one each other in an antagonist-like manner, ii) 5-HT2A antagonists are indirect positive allosteric modulator of mGlu2/3 autoreceptors controlling glutamate exocytosis.
Collapse
Affiliation(s)
- Guendalina Olivero
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, Viale Cembrano 4, 16148, Genoa, Italy
| | - Massimo Grilli
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, Viale Cembrano 4, 16148, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV, 16132, Genoa, Italy
| | - Matteo Vergassola
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, Viale Cembrano 4, 16148, Genoa, Italy
| | - Tommaso Bonfiglio
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, Viale Cembrano 4, 16148, Genoa, Italy
| | - Cristina Padolecchia
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, Viale Cembrano 4, 16148, Genoa, Italy
| | - Beatrice Garrone
- Angelini RR&D (Research, Regulatory & Development) - Angelini S.p.A., Piazzale della Stazione Snc, 00071, S. Palomba-Pomezia (Rome), Italy
| | - Francesco Paolo Di Giorgio
- Angelini RR&D (Research, Regulatory & Development) - Angelini S.p.A., Piazzale della Stazione Snc, 00071, S. Palomba-Pomezia (Rome), Italy
| | - Serena Tongiani
- Angelini RR&D (Research, Regulatory & Development) - Angelini S.p.A., Piazzale della Stazione Snc, 00071, S. Palomba-Pomezia (Rome), Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149, Genoa, Italy
| | - Mario Marchi
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, Viale Cembrano 4, 16148, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV, 16132, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, Viale Cembrano 4, 16148, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV, 16132, Genoa, Italy.
| |
Collapse
|
32
|
Wang CC, Kuo JR, Huang SK, Wang SJ. Metabotropic glutamate 7 receptor agonist AMN082 inhibits glutamate release in rat cerebral cortex nerve terminal. Eur J Pharmacol 2018; 823:11-18. [PMID: 29378190 DOI: 10.1016/j.ejphar.2018.01.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/18/2018] [Accepted: 01/23/2018] [Indexed: 12/21/2022]
Abstract
AMN082 is a selective metabotropic glutamate mGlu7 receptor agonist reported to exhibit antidepressant activity. Considering that excessive glutamate release is involved in the pathogenesis of depression, the effect of N,N'-dibenzyhydryl-ethane-1,2-diamine dihydrochloride (AMN082) on glutamate release in rat cerebrocortical nerve terminals and the possible underlying mechanism were investigated. In this study, we observed here that AMN082 inhibited 4-aminopyridine-evoked glutamate release and this phenomenon was blocked by the metabotropic glutamate mGlu7 receptor antagonist MMPIP. Moreover, western blot analysis and immunocytochemistry confirmed the presence of presynaptic metabotropic glutamate mGlu7 receptor proteins. The effect of AMN082 on the 4-aminopyridine-evoked release of glutamate was prevented by chelating the extracellular Ca2+ ions and the vesicular transporter inhibitor; however, the effect of AMN082 was unaffected by the glutamate transporter inhibitor. AMN082 reduced the elevation of 4-aminopyridine-evoked intrasynaptosomal Ca2+ concentration, but did not alter the synaptosomal membrane potential. In the presence of the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker, the adenylate cyclase inhibitor, and the protein kinase A inhibitor, the action of AMN082 on the 4-aminopyridine-evoked glutamate release was markedly reduced. These results suggest that the activation of the metabotropic glutamate mGlu7 receptors by AMN082 reduces adenylate cyclase/protein kinase A activation, which subsequently reduces the entry of Ca2+ through voltage-dependent Ca2+ channels and decreases evoked glutamate release. Additionally, fluoxetine, a clinically effective antidepressant, completely occluded the inhibitory effect of AMN082 on glutamate release, thus indicating the existence of a common intracellular mechanism for these two compounds to inhibit glutamate release from the cerebrocortical nerve terminals.
Collapse
Affiliation(s)
- Che Chuan Wang
- Department of Neurology, Chi Mei Medical Center, Tainan, Taiwan, ROC; Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan, ROC
| | - Jinn Rung Kuo
- Department of Neurology, Chi Mei Medical Center, Tainan, Taiwan, ROC; Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan, ROC
| | - Shu Kuei Huang
- Department of Anesthesiology, Far-Eastern Memorial Hospital, Pan-Chiao District, New Taipei City 22060, Taiwan, ROC
| | - Su Jane Wang
- School of Medicine, Fu Jen Catholic University, No. 510, Chung-Cheng Rd., Hsin-Chuang, New Taipei 24205, Taiwan, ROC; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan, ROC.
| |
Collapse
|
33
|
Bonfiglio T, Olivero G, Vergassola M, Di Cesare Mannelli L, Pacini A, Iannuzzi F, Summa M, Bertorelli R, Feligioni M, Ghelardini C, Pittaluga A. Environmental training is beneficial to clinical symptoms and cortical presynaptic defects in mice suffering from experimental autoimmune encephalomyelitis. Neuropharmacology 2018; 145:75-86. [PMID: 29402503 DOI: 10.1016/j.neuropharm.2018.01.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 01/19/2018] [Accepted: 01/20/2018] [Indexed: 01/08/2023]
Abstract
The effect of "prophylactic" environmental stimulation on clinical symptoms and presynaptic defects in mice suffering from the experimental autoimmune encephalomyelitis (EAE) at the acute stage of disease (21 ± 1 days post immunization, d.p.i.) was investigated. In EAE mice raised in an enriched environment (EE), the clinical score was reduced when compared to EAE mice raised in standard environment (SE).Concomitantly, gain of weight and increased spontaneous motor activity and curiosity were observed, suggesting increased well-being in mice. Impaired glutamate exocytosis and cyclic adenosine monophosphate (cAMP) production in cortical terminals of SE-EAE mice were evident at 21 ± 1 d.p.i.. Differently, the 12 mM KCl-evoked glutamate exocytosis from cortical synaptosomes of EE-EAE mice was comparable to that observed in SE and EE-control mice, but significantly higher than that in SE-EAE mice. Similarly, the 12 mM KCl-evoked cAMP production in EE-EAE mice cortical synaptosomes recovered to the level observed in SE and EE-control mice. MUNC-18 and SNAP25 contents, but not Syntaxin-1a and Synaptotagmin 1 levels, were increased in cortical synaptosomes from EE-EAE mice when compared to SE-EAE mice. Circulating IL-1β was increased in the spinal cord, but not in the cortex, of SE-EAE mice, and it did not recover in EE-EAE mice. Inflammatory infiltrates were reduced in the cortex but not in the spinal cord of EE-EAE mice. Demyelination was observed in the spinal cord; EE significantly diminished it. We conclude that "prophylactic" EE is beneficial to synaptic derangements and preserves glutamate transmission in the cortex of EAE mice. This article is part of the Special Issue entitled "Neurobiology of Environmental Enrichment".
Collapse
Affiliation(s)
- T Bonfiglio
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - G Olivero
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - M Vergassola
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - L Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Italy
| | - A Pacini
- Department of Experimental and Clinical Medicine, DMSC, Section of Anatomy and Histology, University of Florence, Italy
| | - F Iannuzzi
- EBRI-European Brain Research Institute, Rome, Italy
| | - M Summa
- D3. PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy
| | - R Bertorelli
- D3. PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy
| | - M Feligioni
- EBRI-European Brain Research Institute, Rome, Italy; Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - C Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Italy
| | - A Pittaluga
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy; Centre of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV, 16132, Genoa, Italy.
| |
Collapse
|
34
|
Dammann F, Kirschstein T, Guli X, Müller S, Porath K, Rohde M, Tokay T, Köhling R. Bidirectional shift of group III metabotropic glutamate receptor-mediated synaptic depression in the epileptic hippocampus. Epilepsy Res 2017; 139:157-163. [PMID: 29224956 DOI: 10.1016/j.eplepsyres.2017.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/08/2017] [Accepted: 12/02/2017] [Indexed: 01/20/2023]
Abstract
A common function of group III metabotropic glutamate receptors (mGluRs) located at the presynaptic site of a glutamatergic synapse is synaptic depression. Here, we studied synaptic depression mediated by group III mGluR activation at Schaffer collateral-CA1 (SC-CA1) synapses and associational-commissural-CA3 (AC-CA3) synapses by recording field excitatory postsynaptic potentials in the in vitro brain slice preparation. In order to gauge the impact of synaptic depression in chronically epileptic tissue, we compared rats after pilocarpine-induced status epilepticus (post-SE) with control animals. We observed that synaptic transmission at control AC-CA3 synapses was sensitive to the group III mGluR agonist L-AP4 (10μM), while there was no effect of this compound at SC-CA1 synapses in the same tissue. In contrast, synaptic depression at AC-CA3 synapses by L-AP4 was lost in chronically epileptic tissue, and we found a significant synaptic depression at SC-CA1 synapses in post-SE tissue by L-AP4 and by the mGluR8-selective agonist DCPG. The depression by L-AP4 and DCPG in CA1 was also demonstrated in immature control tissue suggesting developmental down-regulation of mGluR8 at this synapse as well as re-appearance of this isoform under pathological conditions. Quantitative real-time RT-PCR was used to identify mGluR isoforms and to assess their transcriptional changes in post-SE tissue. These analyses revealed down-regulation of mGluR4 and mGluR6 at AC-CA3 and up-regulation of mGluR8 at SC-CA1 synapses. We conclude that group III mGluR-mediated synaptic depression is differentially altered in chronically epileptic tissue by a bidirectional shift of the transcriptional level.
Collapse
Affiliation(s)
- Fabian Dammann
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.
| | - Xiati Guli
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Steffen Müller
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Katrin Porath
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Marco Rohde
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Tursonjan Tokay
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| |
Collapse
|
35
|
Olivero G, Bonfiglio T, Vergassola M, Usai C, Riozzi B, Battaglia G, Nicoletti F, Pittaluga A. Immuno-pharmacological characterization of group II metabotropic glutamate receptors controlling glutamate exocytosis in mouse cortex and spinal cord. Br J Pharmacol 2017; 174:4785-4796. [PMID: 28967122 PMCID: PMC5727332 DOI: 10.1111/bph.14061] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND PURPOSE We recently proposed the existence of mGlu3 -preferring autoreceptors in spinal cord terminals and of mGlu2 -preferring autoreceptors in cortical terminals. This study aims to verify our previous conclusions and to extend their pharmacological characterization. EXPERIMENTAL APPROACH We studied the effect of LY566332, an mGlu2 receptor positive allosteric modulator (PAM), and of LY2389575, a selective mGlu3 receptor negative allosteric (NAM) modulator, on the mGlu2/3 agonist LY379268-mediated inhibition of glutamate exocytosis [measured as KCl-evoked release of preloaded [3 H]-D-aspartate]. The mGlu2 PAM BINA and the mGlu3 NAM ML337, as well as selective antibodies recognizing the N-terminal of the receptor proteins, were used to confirm the pharmacological characterization of the native receptors. KEY RESULTS Cortical synaptosomes possess LY566332-sensitive autoreceptors that are slightly, although significantly, susceptible to LY2389575. In contrast, LY566332-insensitive and LY2389575-sensitive autoreceptors are present in spinal cord terminals. BINA and ML337 mimicked LY566332 and LY2389575, respectively, in controlling LY379268-mediated inhibition of glutamate exocytosis from both cortical and spinal cord synaptosomes. Incubation of cortical synaptosomes with anti-mGlu2 antibody prevented the LY379268-induced inhibition of glutamate exocytosis, and this response was partially reduced by the anti-mGlu3 antibody. Incubation of spinal cord synaptosomes with the anti-mGlu3 antibody abolished LY379268-mediated reduction of glutamate exocytosis from these terminals, while the anti-mGlu2 antibody was inactive. Western blot analysis and confocal microscopy data were largely consistent with these functional observations. CONCLUSIONS AND IMPLICATIONS We confirmed that mGlu3 -preferring autoreceptors exist in spinal cord terminals. Differently, cortical glutamatergic terminals possess mGlu2 /mGlu3 heterodimers, whose inhibitory effect is largely mediated by mGlu2 receptors.
Collapse
Affiliation(s)
| | | | | | - Cesare Usai
- National Research CouncilInstitute of BiophysicsGenoaItaly
| | | | | | - Ferdinando Nicoletti
- I.R.C.C.S. Neuromed, Località CamerellePozzilliItaly
- Department of Physiology and PharmacologySapienza UniversityRomeItaly
| | - Anna Pittaluga
- Department of Pharmacy, DiFARUniversity of GenoaGenoaItaly
- Centre of Excellence for Biomedical ResearchUniversity of GenoaGenoaItaly
| |
Collapse
|
36
|
Pittaluga A. CCL5-Glutamate Cross-Talk in Astrocyte-Neuron Communication in Multiple Sclerosis. Front Immunol 2017; 8:1079. [PMID: 28928746 PMCID: PMC5591427 DOI: 10.3389/fimmu.2017.01079] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/18/2017] [Indexed: 12/17/2022] Open
Abstract
The immune system (IS) and the central nervous system (CNS) are functionally coupled, and a large number of endogenous molecules (i.e., the chemokines for the IS and the classic neurotransmitters for the CNS) are shared in common between the two systems. These interactions are key elements for the elucidation of the pathogenesis of central inflammatory diseases. In recent years, evidence has been provided supporting the role of chemokines as modulators of central neurotransmission. It is the case of the chemokines CCL2 and CXCL12 that control pre- and/or post-synaptically the chemical transmission. This article aims to review the functional cross-talk linking another endogenous pro-inflammatory factor released by glial cells, i.e., the chemokine Regulated upon Activation Normal T-cell Expressed and Secreted (CCL5) and the principal neurotransmitter in CNS (i.e., glutamate) in physiological and pathological conditions. In particular, the review discusses preclinical data concerning the role of CCL5 as a modulator of central glutamatergic transmission in healthy and demyelinating disorders. The CCL5-mediated control of glutamate release at chemical synapses could be relevant either to the onset of psychiatric symptoms that often accompany the development of multiple sclerosis (MS), but also it might indirectly give a rationale for the progression of inflammation and demyelination. The impact of disease-modifying therapies for the cure of MS on the endogenous availability of CCL5 in CNS will be also summarized. We apologize in advance for omission in our coverage of the existing literature.
Collapse
Affiliation(s)
- Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
37
|
Bonfiglio T, Olivero G, Merega E, Di Prisco S, Padolecchia C, Grilli M, Milanese M, Di Cesare Mannelli L, Ghelardini C, Bonanno G, Marchi M, Pittaluga A. Prophylactic versus Therapeutic Fingolimod: Restoration of Presynaptic Defects in Mice Suffering from Experimental Autoimmune Encephalomyelitis. PLoS One 2017; 12:e0170825. [PMID: 28125677 PMCID: PMC5268435 DOI: 10.1371/journal.pone.0170825] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 01/11/2017] [Indexed: 11/18/2022] Open
Abstract
Fingolimod, the first oral, disease-modifying therapy for MS, has been recently proposed to modulate glutamate transmission in the central nervous system (CNS) of mice suffering from Experimental Autoimmune Encephalomyelitis (EAE) and in MS patients. Our study aims at investigating whether oral fingolimod recovers presynaptic defects that occur at different stages of disease in the CNS of EAE mice. In vivo prophylactic (0.3 mg/kg for 14 days, from the 7th day post immunization, d.p.i, the drug dissolved in the drinking water) fingolimod significantly reduced the clinical symptoms and the anxiety-related behaviour in EAE mice. Spinal cord inflammation, demyelination and glial cell activation are markers of EAE progression. These signs were ameliorated following oral fingolimod administration. Glutamate exocytosis was shown to be impaired in cortical and spinal cord terminals isolated from EAE mice at 21 ± 1 d.p.i., while GABA alteration emerged only at the spinal cord level. Prophylactic fingolimod recovered these presynaptic defects, restoring altered glutamate and GABA release efficiency. The beneficial effect occurred in a dose-dependent, region-specific manner, since lower (0.1-0.03 mg/kg) doses restored, although to a different extent, synaptic defects in cortical but not spinal cord terminals. A delayed reduction of glutamate, but not of GABA, exocytosis was observed in hippocampal terminals of EAE mice at 35 d.p.i. Therapeutic (0.3 mg/kg, from 21 d.p.i. for 14 days) fingolimod restored glutamate exocytosis in the cortex and in the hippocampus of EAE mice at 35 ± 1 d.p.i. but not in the spinal cord, where also GABAergic defects remained unmodified. These results improve our knowledge of the molecular events accounting for the beneficial effects elicited by fingolimod in demyelinating disorders.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Cerebral Cortex/drug effects
- Cerebral Cortex/immunology
- Cerebral Cortex/pathology
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Exocytosis/drug effects
- Female
- Fingolimod Hydrochloride/pharmacology
- Glutamic Acid/metabolism
- Glutamic Acid/pharmacology
- Hippocampus/drug effects
- Hippocampus/immunology
- Hippocampus/pathology
- Immunosuppressive Agents/pharmacology
- Mice
- Mice, Inbred C57BL
- Neuroglia/drug effects
- Neuroglia/immunology
- Neuroglia/pathology
- Organ Specificity
- Spinal Cord/drug effects
- Spinal Cord/immunology
- Spinal Cord/pathology
- Synapses/drug effects
- Synapses/immunology
- Synapses/pathology
- gamma-Aminobutyric Acid/metabolism
- gamma-Aminobutyric Acid/pharmacology
Collapse
Affiliation(s)
- Tommaso Bonfiglio
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Guendalina Olivero
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Elisa Merega
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Silvia Di Prisco
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Cristina Padolecchia
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Massimo Grilli
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Marco Milanese
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology section, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology section, University of Florence, Florence, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Mario Marchi
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
38
|
Di Prisco S, Olivero G, Merega E, Bonfiglio T, Marchi M, Pittaluga A. CXCR4 and NMDA Receptors Are Functionally Coupled in Rat Hippocampal Noradrenergic and Glutamatergic Nerve Endings. J Neuroimmune Pharmacol 2016; 11:645-656. [PMID: 27147258 DOI: 10.1007/s11481-016-9677-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 04/25/2016] [Indexed: 01/31/2023]
Abstract
Previous studies had shown that the HIV-1 capsidic glycoprotein gp120 (strain IIIB) modulates presynaptic release-regulating NMDA receptors on noradrenergic and glutamatergic terminals. This study aims to assess whether the chemokine CXC4 receptors (CXCR4s) has a role in the gp120-mediated effects. The effect of CXCL12, the endogenous ligand at CXCR4, on the NMDA-mediated releasing activity was therefore investigated. Rat hippocampal synaptosomes were preloaded with [3H]noradrenaline ([3H]NA) or [3H]D-aspartate ([3H]D-Asp) and acutely exposed to CXCL12, to NMDA or to both agonists. CXCL12, inactive on its own, facilitated the NMDA-evoked tritium release. The NMDA antagonist MK-801 abolished the NMDA/CXCL12-evoked tritium release of both radiolabelled tracers, while the CXCR4 antagonist AMD 3100 halved it, suggesting that rat hippocampal nerve endings possess presynaptic release-regulating CXCR4 receptors colocalized with NMDA receptors. Accordingly, Western blot analysis confirmed the presence of CXCR4 proteins in synaptosomal plasmamembranes. In both synaptosomal preparations, CXCL12-induced facilitation of NMDA-mediated release was dependent upon PLC-mediated src-induced events leading to mobilization of Ca2+ from intraterminal IP3-sensitive stores Finally, the gp120-induced facilitation of NMDA-mediated release of [3H]NA and [3H]D-Asp was prevented by AMD 3100. We propose that CXCR4s are functionally coupled to NMDA receptors in rat hippocampal noradrenergic and glutamatergic terminals and account for the gp120-induced modulation of the NMDA-mediated central effects. The NMDA/CXCR4 cross-talk could have a role in the neuropsychiatric symptoms often observed in HIV-1 positive patients.
Collapse
Affiliation(s)
- Silvia Di Prisco
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Guendalina Olivero
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Elisa Merega
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Tommaso Bonfiglio
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy
| | - Mario Marchi
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV, 16132, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, 16148, Genoa, Italy. .,Center of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV, 16132, Genoa, Italy.
| |
Collapse
|
39
|
Di Prisco S, Merega E, Bonfiglio T, Olivero G, Cervetto C, Grilli M, Usai C, Marchi M, Pittaluga A. Presynaptic, release-regulating mGlu2 -preferring and mGlu3 -preferring autoreceptors in CNS: pharmacological profiles and functional roles in demyelinating disease. Br J Pharmacol 2016; 173:1465-77. [PMID: 26791341 DOI: 10.1111/bph.13442] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 01/08/2016] [Accepted: 01/17/2016] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE Presynaptic, release-regulating metabotropic glutamate 2 and 3 (mGlu2/3) autoreceptors exist in the CNS. They represent suitable targets for therapeutic approaches to central diseases that are typified by hyperglutamatergicity. The availability of specific ligands able to differentiate between mGlu2 and mGlu3 subunits allows us to further characterize these autoreceptors. In this study we investigated the pharmacological profile of mGlu2/3 receptors in selected CNS regions and evaluated their functions in mice with experimental autoimmune encephalomyelitis (EAE). EXPERIMENTAL APPROACH The comparative analysis of presynaptic mGlu2/3 autoreceptors was performed by determining the effect of selective mGlu2/3 receptor agonist(s) and antagonist(s) on the release of [(3)H]-D-aspartate from cortical and spinal cord synaptosomes in superfusion. In EAE mice, mGlu2/3 autoreceptor-mediated release functions were investigated and effects of in vivo LY379268 administration on impaired glutamate release examined ex vivo. KEY RESULTS Western blot analysis and confocal microscopy confirmed the presence of presynaptic mGlu2/3 receptor proteins. Cortical synaptosomes possessed LY541850-sensitive, NAAG-insensitive autoreceptors having low affinity for LY379268, while LY541850-insensitive, NAAG-sensitive autoreceptors with high affinity for LY379268 existed in spinal cord terminals. In EAE mice, mGlu2/3 autoreceptors completely lost their inhibitory activity in cortical, but not in spinal cord synaptosomes. In vivo LY379268 administration restored the glutamate exocytosis capability in spinal cord but not in cortical terminals in EAE mice. CONCLUSIONS AND IMPLICATIONS We propose the existence of mGlu2-preferring and mGlu3-preferring autoreceptors in mouse cortex and spinal cord respectively. The mGlu3 -preferring autoreceptors could represent a target for new pharmacological approaches for treating demyelinating diseases.
Collapse
Affiliation(s)
- Silvia Di Prisco
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Elisa Merega
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Tommaso Bonfiglio
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Guendalina Olivero
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Chiara Cervetto
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Massimo Grilli
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council, Genoa, Italy
| | - Mario Marchi
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
40
|
MMPIP, an mGluR7-selective negative allosteric modulator, alleviates pain and normalizes affective and cognitive behavior in neuropathic mice. Pain 2016; 156:1060-1073. [PMID: 25760470 DOI: 10.1097/j.pain.0000000000000150] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This study investigated the effects of a single administration of 6-(4-methoxyphenyl)-5-methyl-3-pyridinyl-4-isoxazolo[4,5-c]pyridin-4(5H)-one (MMPIP), a negative allosteric modulator (NAM) of metabotropic glutamate receptor 7 (mGluR7), on pain and on affective and cognitive behavior in neuropathic mice. The activity of pyramidal neurons in the prelimbic cortex (PLC), which respond to stimulation of the basolateral amygdala (BLA) with either excitation or inhibition, was also investigated. The spared nerve injury (SNI) of the sciatic nerve induced, 14 days after surgery, thermal hyperalgesia and mechanical allodynia, reduced open-arm choice in the elevated plus-maze, increased time of immobility in the tail suspension, and increased digging and burying in the marble burying test. Cognitive performance was also significantly compromised in the SNI mice. Spared nerve injury induced phenotypic changes on pyramidal neurons of the PLC; excitatory responses increased, whereas inhibitory responses decreased after BLA stimulation. mGluR7 expression, mainly associated with vesicular glutamate transporter, increased in the hippocampus and decreased in the BLA, PLC, and dorsal raphe in SNI mice. MMPIP increased thermal and mechanical thresholds and open-arm choice. It reduced the immobility in the tail suspension test and the number of marbles buried and of digging events in the marble burying test. MMPIP also improved cognitive performance and restored the balance between excitatory and inhibitory responses of PLC neurons in SNI mice. 7-hydroxy-3-(4-iodophenoxy)-4H-chromen-4-one, XAP044, another selective mGluR7 NAM, reproduced the effects of MMPIP on thermal hyperalgesia, mechanical allodynia, tail suspension, and marble burying test. Altogether, these findings show that mGluR7 NAMs reduce pain responses and affective/cognitive impairments in neuropathic pain conditions.
Collapse
|
41
|
Metabotropic Glutamate Receptor Subtype 7 in the Bed Nucleus of the Stria Terminalis is Essential for Intermale Aggression. Neuropsychopharmacology 2016; 41:726-35. [PMID: 26149357 PMCID: PMC4707819 DOI: 10.1038/npp.2015.198] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/23/2015] [Accepted: 06/29/2015] [Indexed: 12/25/2022]
Abstract
Metabotropic glutamate receptor subtype 7 (mGluR7) is a member of group III mGluRs, which localize to the presynaptic active zones of the mammalian central nervous system. Although histological, genetic, and electrophysiological studies ensure the importance of mGluR7, its roles in behavior and physiology remain largely unknown. Using a resident-intruder paradigm, we found a severe reduction in intermale aggressive behavior in mGluR7 knockout (KO) mice. We also found alterations in other social behaviors in male mGluR7 KO mice, including sexual behavior toward male intruders. Because olfaction is critical for rodent social behavior, including aggression, we performed an olfaction test, finding that mGluR7 KO mice failed to show interest in the smell of male urine. To clarify the olfactory deficit, we then exposed mice to urine and analyzed c-Fos-immunoreactivity, discovering a remarkable reduction in neural activity in the bed nucleus of the stria terminalis (BNST) of mGluR7 KO mice. Finally, intra-BNST administration of the mGluR7-selective antagonist 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazolo[4,5-c]pyridin-4(5H)-one (MMPIP) also reproduced the phenotype of mGluR7 KO mice, including reduced aggression and altered social interaction. Thus mGluR7 may work as an 'enhancer of neural activity' in the BNST and is important for intermale aggression. Our findings demonstrate that mGluR7 is essential for social behavior and innate behavior. Our study on mGluR7 in the BNST will shed light on future therapies for emotional disorders in humans.
Collapse
|
42
|
Peterlik D, Flor PJ, Uschold-Schmidt N. The Emerging Role of Metabotropic Glutamate Receptors in the Pathophysiology of Chronic Stress-Related Disorders. Curr Neuropharmacol 2016; 14:514-39. [PMID: 27296643 PMCID: PMC4983752 DOI: 10.2174/1570159x13666150515234920] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 04/04/2015] [Accepted: 05/12/2015] [Indexed: 12/28/2022] Open
Abstract
Chronic stress-related psychiatric conditions such as anxiety, depression, and alcohol abuse are an enormous public health concern. The etiology of these pathologies is complex, with psychosocial stressors being among the most frequently discussed risk factors. The brain glutamatergic neurotransmitter system has often been found involved in behaviors and pathophysiologies resulting from acute stress and fear. Despite this, relatively little is known about the role of glutamatergic system components in chronic psychosocial stress, neither in rodents nor in humans. Recently, drug discovery efforts at the metabotropic receptor subtypes of the glutamatergic system (mGlu1-8 receptors) led to the identification of pharmacological tools with emerging potential in psychiatric conditions. But again, the contribution of individual mGlu subtypes to the manifestation of physiological, molecular, and behavioral consequences of chronic psychosocial stress remains still largely unaddressed. The current review will describe animal models typically used to analyze acute and particularly chronic stress conditions, including models of psychosocial stress, and there we will discuss the emerging roles for mGlu receptor subtypes. Indeed, accumulating evidence indicates relevance and potential therapeutic usefulness of mGlu2/3 ligands and mGlu5 receptor antagonists in chronic stress-related disorders. In addition, a role for further mechanisms, e.g. mGlu7-selective compounds, is beginning to emerge. These mechanisms are important to be analyzed in chronic psychosocial stress paradigms, e.g. in the chronic subordinate colony housing (CSC) model. We summarize the early results and discuss necessary future investigations, especially for mGlu5 and mGlu7 receptor blockers, which might serve to suggest improved therapeutic strategies to treat stress-related disorders.
Collapse
Affiliation(s)
| | - Peter J Flor
- Faculty of Biology and Preclinical Medicine, University of Regensburg, D-93053 Regensburg, Germany.
| | - Nicole Uschold-Schmidt
- Faculty of Biology and Preclinical Medicine, University of Regensburg, D-93053 Regensburg, Germany.
| |
Collapse
|
43
|
Bragina L, Bonifacino T, Bassi S, Milanese M, Bonanno G, Conti F. Differential expression of metabotropic glutamate and GABA receptors at neocortical glutamatergic and GABAergic axon terminals. Front Cell Neurosci 2015; 9:345. [PMID: 26388733 PMCID: PMC4559644 DOI: 10.3389/fncel.2015.00345] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/18/2015] [Indexed: 12/04/2022] Open
Abstract
Metabotropic glutamate (Glu) receptors (mGluRs) and GABAB receptors are highly expressed at presynaptic sites. To verify the possibility that the two classes of metabotropic receptors contribute to axon terminals heterogeneity, we studied the localization of mGluR1α, mGluR5, mGluR2/3, mGluR7, and GABAB1 in VGLUT1-, VGLUT2-, and VGAT- positive terminals in the cerebral cortex of adult rats. VGLUT1-positive puncta expressed mGluR1α (∼5%), mGluR5 (∼6%), mGluR2/3 (∼22%), mGluR7 (∼17%), and GABAB1 (∼40%); VGLUT2-positive terminals expressed mGluR1α (∼10%), mGluR5 (∼11%), mGluR2/3 (∼20%), mGluR7 (∼28%), and GABAB1 (∼25%); whereas VGAT-positive puncta expressed mGluR1α (∼27%), mGluR5 (∼24%), mGluR2/3 (∼38%), mGluR7 (∼31%), and GABAB1 (∼19%). Control experiments ruled out the possibility that postsynaptic mGluRs and GABAB1 might have significantly biased our results. We also performed functional assays in synaptosomal preparations, and showed that all agonists modify Glu and GABA levels, which return to baseline upon exposure to antagonists. Overall, these findings indicate that mGluR1α, mGluR5, mGluR2/3, mGluR7, and GABAB1 expression differ significantly between glutamatergic and GABAergic axon terminals, and that the robust expression of heteroreceptors may contribute to the homeostatic regulation of the balance between excitation and inhibition.
Collapse
Affiliation(s)
- Luca Bragina
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
- Center for Neurobiology of Aging, Istituto Nazionale di Riposo e Cura per Anziani – Istituto di Ricovero e Cura a Carattere ScientificoAncona, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of GenoaGenoa, Italy
| | - Silvia Bassi
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of GenoaGenoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of GenoaGenoa, Italy
- Center of Excellence for Biomedical Research, University of GenoaGenoa, Italy
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
- Center for Neurobiology of Aging, Istituto Nazionale di Riposo e Cura per Anziani – Istituto di Ricovero e Cura a Carattere ScientificoAncona, Italy
- Fondazione di Medicina Molecolare, Università Politecnica delle MarcheAncona, Italy
| |
Collapse
|
44
|
Activation of Metabotropic Glutamate Receptor 7 Is Required for Induction of Long-Term Potentiation at SC-CA1 Synapses in the Hippocampus. J Neurosci 2015; 35:7600-15. [PMID: 25972184 DOI: 10.1523/jneurosci.4543-14.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Of the eight metabotropic glutamate (mGlu) receptor subtypes, only mGlu7 is expressed presynaptically at the Schaffer collateral (SC)-CA1 synapse in the hippocampus in adult animals. Coupled with the inhibitory effects of Group III mGlu receptor agonists on transmission at this synapse, mGlu7 is thought to be the predominant autoreceptor responsible for regulating glutamate release at SC terminals. However, the lack of mGlu7-selective pharmacological tools has hampered direct testing of this hypothesis. We used a novel, selective mGlu7-negative allosteric modulator (NAM), ADX71743, and a newly described Group III mGlu receptor agonist, LSP4-2022, to elucidate the role of mGlu7 in modulating transmission in hippocampal area CA1 in adult C57BL/6J male mice. Interestingly, although mGlu7 agonists inhibit SC-CA1 EPSPs, we found no evidence for activation of mGlu7 by stimulation of SC-CA1 afferents. However, LSP4-2022 also reduced evoked monosynaptic IPSCs in CA1 pyramidal cells and, in contrast to its effect on SC-CA1 EPSPs, ADX71743 reversed the ability of high-frequency stimulation of SC afferents to reduce IPSC amplitudes. Furthermore, blockade of mGlu7 prevented induction of LTP at the SC-CA1 synapse and activation of mGlu7 potentiated submaximal LTP. Together, these data suggest that mGlu7 serves as a heteroreceptor at inhibitory synapses in area CA1 and that the predominant effect of activation of mGlu7 by stimulation of glutamatergic afferents is disinhibition, rather than reduced excitatory transmission. Furthermore, this mGlu7-mediated disinhibition is required for induction of LTP at the SC-CA1 synapse, suggesting that mGlu7 could serve as a novel therapeutic target for treatment of cognitive disorders.
Collapse
|
45
|
Stensrud MJ, Sogn CJ, Gundersen V. Immunogold characteristics of VGLUT3-positive GABAergic nerve terminals suggest corelease of glutamate. J Comp Neurol 2015; 523:2698-713. [PMID: 26010578 DOI: 10.1002/cne.23811] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 05/18/2015] [Accepted: 05/18/2015] [Indexed: 12/20/2022]
Abstract
There is compelling evidence that glutamate can act as a cotransmitter in the mammalian brain. Interestingly, the third vesicular glutamate transporter (VGLUT3) is primarily found in neurons that were anticipated to be nonglutamatergic. Whereas the function of VGLUT3 in acetylcholinergic and serotoninergic neurons has been elucidated, the role of VGLUT3 in neurons releasing gamma-aminobutyric acid (GABA) is not settled. We have previously shown that VGLUT3 is found together with the vesicular GABA transporter (VIAAT) on synaptic vesicle membranes in the hippocampus. Now we provide novel electron microscopic data from the rat hippocampus suggesting that glutamate is enriched in inhibitory nerve terminals containing VGLUT3 compared to those lacking VGLUT3. The opposite was found for GABA; VGLUT3-positive inhibitory terminals contained lower density of GABA labeling compared to VGLUT3-negative inhibitory terminals. In addition, semiquantitative confocal immunofluorescence showed that N-methyl-D-aspartate (NMDA)-receptor labeling was present more frequently in VGLUT3-positive/VIAAT-positive synapses versus in VGLUT3-negative/VIAAT-positive synapses. Electron microscopic immunogold data further suggest that NMDA receptors are enriched in VGLUT3 containing inhibitory terminals. Our data reveal new chemical characteristics of a subset of GABAergic interneurons in the hippocampus. The analyses suggest that glutamate is coreleased with GABA from hippocampal basket cell-synapses to act on NMDA receptors.
Collapse
Affiliation(s)
- Mats Julius Stensrud
- Department of Anatomy and Healthy Brain Ageing Centre Regional Research Network, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Carl Johan Sogn
- Department of Anatomy and Healthy Brain Ageing Centre Regional Research Network, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Vidar Gundersen
- Department of Anatomy and Healthy Brain Ageing Centre Regional Research Network, Institute of Basic Medical Sciences, University of Oslo, Norway.,Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
46
|
Merega E, Prisco SD, Severi P, Kalfas F, Pittaluga A. Antibody/receptor protein immunocomplex in human and mouse cortical nerve endings amplifies complement-induced glutamate release. Neurosci Lett 2015; 600:50-5. [PMID: 26049008 DOI: 10.1016/j.neulet.2015.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/31/2015] [Accepted: 06/01/2015] [Indexed: 10/23/2022]
Abstract
Previous studies have demonstrated that complement alone releases glutamate from human and mouse cortical terminals in an antibody-independent manner. In order to expand our knowledge on complement-mediated effects, we investigated whether the presence of an antigen-antibody complex in synaptosomal plasmamembranes could also trigger complement-induced functional responses that might affect neurotransmitter release. To this aim, we focused on the chemokine 5 receptor (CCR5) expressed in human and mouse cortical glutamate terminals, whose activation by CCL5 elicits [(3)H]D-aspartate ([(3)H]D-ASP) release. Preincubating synaptosomes with an antibody recognizing the NH2 terminus of the CCR5 protein (anti-NH2-CCR5 antibody) abolished the CCL5-induced [(3)H]D-ASP release. Similarly, enriching synaptosomes with an antibody recognizing the COOH terminus of CCR5 (anti-COOH-CCR5 antibody) prevented the CCL5-induced [(3)H]D-ASP release. The antagonist-like activity of the anti-NH2-CCR5 antibody turned to facilitation when anti-NH2-CCR5-treated synaptosomes were exposed to complement. In these terminals, the releasing effect was significantly higher than that elicited by complement in untreated synaptosomes. On the contrary, the complement-induced [(3)H]D-ASP release from anti-COOH-CCR5 antibody-entrapped synaptosomes did not differ from that from untreated synaptosomes. Preincubating synaptosomes with anti-beta tubulin III antibody, used as negative control, neither prevented the CCL5-induced releasing effect nor it amplified the complement-induced [(3)H]D-ASP release. Finally, serum lacking the C1q protein, i.e. the protein essential to promote the antibody-mediated activation of complement, elicited a comparable [(3)H]D-ASP release from both untreated and anti-NH2-CCR5 antibody-treated synaptosomes. Thus, we propose that antibodies raised against the outer sequence of a receptor protein can trigger the activation of the complement through the classic, C1q-mediated antibody-dependent pathway, which results in an abnormal release of glutamate that could be deleterious to central nervous system.
Collapse
Affiliation(s)
- Elisa Merega
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Silvia Di Prisco
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Paolo Severi
- Division of Neurosurgery, Galliera Hospital, Genoa, Italy
| | - Fotios Kalfas
- Division of Neurosurgery, Galliera Hospital, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.
| |
Collapse
|
47
|
Di Prisco S, Merega E, Lanfranco M, Casazza S, Uccelli A, Pittaluga A. Acute desipramine restores presynaptic cortical defects in murine experimental autoimmune encephalomyelitis by suppressing central CCL5 overproduction. Br J Pharmacol 2014; 171:2457-67. [PMID: 24528439 DOI: 10.1111/bph.12631] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/14/2014] [Accepted: 02/05/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Altered glutamate exocytosis and cAMP production in cortical terminals of experimental autoimmune encephalomyelitis (EAE) mice occur at the early stage of disease (13 days post-immunization, d.p.i.). Neuronal defects were paralleled by overexpression of the central chemokine CCL5 (also known as RANTES), suggesting it has a role in presynaptic impairments. We propose that drugs able to restore CCL5 content to physiological levels could also restore presynaptic defects. Because of its efficacy in controlling CCL5 overexpression, desipramine (DMI) appeared to be a suitable candidate to test our hypothesis. EXPERIMENTAL APPROACH Control and EAE mice at 13 d.p.i. were acutely or chronically administered DMI and monitored for behaviour and clinical scores. Noradrenaline and glutamate release, cAMP, CCL5 and TNF-α production were quantified in cortical synaptosomes and homogenates. Peripheral cytokine production was also determined. KEY RESULTS Noradrenaline exocytosis and α₂ -adrenoeceptor-mediated activity were unmodified in EAE mice at 13 d.p.i. when compared with control. Acute, but not chronic, DMI reduced CCL5 levels in cortical homogenates of EAE mice at 13 d.p.i., but did not affect peripheral IL-17 and TNF-α contents or CCL5 plasma levels. Acute DMI caused a long-lasting restoration of glutamate exocytosis, restored endogenous cAMP production and impeded the shift from inhibition to facilitation of the CCL5-mediated control of glutamate exocytosis. Finally, DMI ameliorated anxiety-related behaviour but not motor activity or severity of clinical signs. CONCLUSIONS We propose DMI as an add-on therapy to normalize neuropsychiatric symptoms in multiple sclerosis patients at the early stage of the disease.
Collapse
Affiliation(s)
- Silvia Di Prisco
- Department of Pharmacy, DiFAR, Pharmacology and Toxicology Section, Genoa, Italy
| | | | | | | | | | | |
Collapse
|
48
|
Orlando R, Borro M, Motolese M, Molinaro G, Scaccianoce S, Caruso A, di Nuzzo L, Caraci F, Matrisciano F, Pittaluga A, Mairesse J, Simmaco M, Nisticò R, Monn JA, Nicoletti F. Levels of the Rab GDP dissociation inhibitor (GDI) are altered in the prenatal restrain stress mouse model of schizophrenia and are differentially regulated by the mGlu2/3 receptor agonists, LY379268 and LY354740. Neuropharmacology 2014; 86:133-44. [PMID: 25063582 DOI: 10.1016/j.neuropharm.2014.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 07/07/2014] [Accepted: 07/09/2014] [Indexed: 11/24/2022]
Abstract
LY379268 and LY354740, two agonists of mGlu2/3 metabotropic glutamate receptors, display different potencies in mouse models of schizophrenia. This differential effect of the two drugs remains unexplained. We performed a proteomic analysis in cultured cortical neurons challenged with either LY379268 or LY354740. Among the few proteins that were differentially influenced by the two drugs, Rab GDP dissociation inhibitor-β (Rab GDIβ) was down-regulated by LY379268 and showed a trend to an up-regulation in response to LY354740. In cultured hippocampal neurons, LY379268 selectively down-regulated the α isoform of Rab GDI. Rab GDI inhibits the activity of the synaptic vesicle-associated protein, Rab3A, and is reduced in the brain of schizophrenic patients. We examined the expression of Rab GDI in mice exposed to prenatal stress ("PRS mice"), which have been described as a putative model of schizophrenia. Rab GDIα protein levels were increased in the hippocampus of PRS mice at postnatal days (PND)1 and 21, but not at PND60. At PND21, PRS mice also showed a reduced depolarization-evoked [(3)H]d-aspartate release in hippocampal synaptosomes. The increase in Rab GDIα levels in the hippocampus of PRS mice was reversed by a 7-days treatment with LY379268 (1 or 10 mg/kg, i.p.), but not by treatment with equal doses of LY354740. These data strengthen the validity of PRS mice as a model of schizophrenia, and show for the first time a pharmacodynamic difference between LY379268 and LY354740 which might be taken into account in an attempt to explain the differential effect of the two drugs across mouse models.
Collapse
Affiliation(s)
- Rosamaria Orlando
- IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy
| | - Marina Borro
- NESMOS Department, Advanced Molecular Diagnostic Unit, Sapienza University, Sant'Andrea Hospital, Rome, Italy
| | | | | | - Sergio Scaccianoce
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | - Alessandra Caruso
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | - Luigi di Nuzzo
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | - Filippo Caraci
- IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy; Department of Educational Sciences, University of Catania, Catania, Italy
| | | | - Anna Pittaluga
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Jerome Mairesse
- Neural Plasticity Team, Université Lille 1, International Associated Laboratory (LIA), France
| | - Maurizio Simmaco
- NESMOS Department, Advanced Molecular Diagnostic Unit, Sapienza University, Sant'Andrea Hospital, Rome, Italy
| | - Robert Nisticò
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | - James A Monn
- Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, Italy; Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy.
| |
Collapse
|
49
|
Merega E, Prisco SD, Lanfranco M, Severi P, Pittaluga A. Complement selectively elicits glutamate release from nerve endings in different regions of mammal central nervous system. J Neurochem 2014; 129:473-83. [DOI: 10.1111/jnc.12650] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 12/29/2013] [Accepted: 01/02/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Elisa Merega
- Department of Pharmacy; Pharmacology and Toxicology Section; University of Genoa; Genoa Italy
| | - Silvia Di Prisco
- Department of Pharmacy; Pharmacology and Toxicology Section; University of Genoa; Genoa Italy
| | | | - Paolo Severi
- Division of Neurosurgery; Galliera Hospital; Genoa Italy
| | - Anna Pittaluga
- Department of Pharmacy; Pharmacology and Toxicology Section; University of Genoa; Genoa Italy
- Center of Excellence for Biomedical Research; University of Genoa; Genoa Italy
| |
Collapse
|
50
|
Iacovelli L, Felicioni M, Nisticò R, Nicoletti F, De Blasi A. Selective regulation of recombinantly expressed mGlu7 metabotropic glutamate receptors by G protein-coupled receptor kinases and arrestins. Neuropharmacology 2013; 77:303-12. [PMID: 24148810 DOI: 10.1016/j.neuropharm.2013.10.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/24/2013] [Accepted: 10/07/2013] [Indexed: 12/24/2022]
Abstract
mGlu7 receptors are coupled to Gi/Go-proteins and activate multiple transduction pathways, including inhibition of adenylyl cyclase activity and stimulation of ERK1/2 and JNK pathways. mGlu7 receptors play an important role in cognition and emotion and are involved in stress-related disorders such as anxiety and depression and in susceptibility to convulsive seizures. In spite of these potential clinical implications, little is known on the mechanisms that regulate mGlu7-receptor signaling. Here we show that mGlu7 receptor-dependent signaling pathways were regulated in a complementary manner by different GRK subtypes, with GRK4 affecting the adenylyl cyclase and the JNK pathways, and GRK2 selectively affecting the ERK1/2 pathway. Additionally we found that the two isoforms of non-visual arrestins, i.e. β-arrestin1 and β-arrestin2, exerted opposite effects on mGlu7-receptor signaling, with β-arrestin1 positively modulating ERK1/2 and inhibiting JNK, and β-arrestin2 doing the opposite. This represents a remarkable example of "reciprocal regulation" of receptor signaling by the two isoforms of β-arrestin. Finally we found that β-arrestin1 amplified mGlu7 receptor-dependent ERK1/2 activation in response to L-AP4 (an orthosteric agonist), but not in response to AMN082 (an atypical mGlu7-receptor allosteric agonist). The different effect of β-arrestin1 on L-AP4- and AMN082-stimulated ERK1/2 phosphorylation is in line with the emerging concept of β-arrestin-biased agonists. The present study may open new perspectives in elucidating the physio-pathological roles of the mGlu7 receptor and may provide new insights for the possibility to develop specific (biased) agonists that can selectively activate different signaling pathways.
Collapse
Affiliation(s)
- L Iacovelli
- Department of Physiology and Pharmacology "Vittorio Erspamer", University of Rome "Sapienza", P.le Aldo Moro, 5, 00185 Rome, Italy.
| | - M Felicioni
- IRCSS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - R Nisticò
- Department of Physiology and Pharmacology "Vittorio Erspamer", University of Rome "Sapienza", P.le Aldo Moro, 5, 00185 Rome, Italy; IRCSS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - F Nicoletti
- Department of Physiology and Pharmacology "Vittorio Erspamer", University of Rome "Sapienza", P.le Aldo Moro, 5, 00185 Rome, Italy; I.N.M. Neuromed, Località Camerelle, Pozzilli, Italy
| | - A De Blasi
- Dept. of Molecular Medicine, University of Rome "Sapienza", V.le Regina Elena 291, 00185 Rome, Italy
| |
Collapse
|