1
|
Munguba H, Gutzeit VA, Srivastava I, Kristt M, Singh A, Vijay A, Arefin A, Thukral S, Broichhagen J, Stujenske JM, Liston C, Levitz J. Projection-Targeted Photopharmacology Reveals Distinct Anxiolytic Roles for Presynaptic mGluR2 in Prefrontal- and Insula-Amygdala Synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575699. [PMID: 38293136 PMCID: PMC10827048 DOI: 10.1101/2024.01.15.575699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Dissecting how membrane receptors regulate neural circuit function is critical for deciphering basic principles of neuromodulation and mechanisms of therapeutic drug action. Classical pharmacological and genetic approaches are not well-equipped to untangle the roles of specific receptor populations, especially in long-range projections which coordinate communication between brain regions. Here we use viral tracing, electrophysiological, optogenetic, and photopharmacological approaches to determine how presynaptic metabotropic glutamate receptor 2 (mGluR2) activation in the basolateral amygdala (BLA) alters anxiety-related behavior. We find that mGluR2-expressing neurons from the ventromedial prefrontal cortex (vmPFC) and posterior insular cortex (pIC) preferentially target distinct cell types and subregions of the BLA to regulate different forms of avoidant behavior. Using projection-specific photopharmacological activation, we find that mGluR2-mediated presynaptic inhibition of vmPFC-BLA, but not pIC-BLA, connections can produce long-lasting decreases in spatial avoidance. In contrast, presynaptic inhibition of pIC-BLA connections decreased social avoidance, novelty-induced hypophagia, and increased exploratory behavior without impairing working memory, establishing this projection as a novel target for the treatment of anxiety disorders. Overall, this work reveals new aspects of BLA neuromodulation with therapeutic implications while establishing a powerful approach for optical mapping of drug action via photopharmacology.
Collapse
Affiliation(s)
- Hermany Munguba
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Vanessa A. Gutzeit
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ipsit Srivastava
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Melanie Kristt
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ashna Singh
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Akshara Vijay
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anisul Arefin
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sonal Thukral
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Joseph M. Stujenske
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Conor Liston
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
2
|
Bagheri J, Fallahnezhad S, Alipour N, Babaloo H, Tahmasebi F, Kheradmand H, Sazegar G, Haghir H. Maternal diabetes decreases the expression of GABA Aα1, GABA B1, and mGlu2 receptors in the visual cortex of male rat neonates. Neurosci Lett 2023; 809:137309. [PMID: 37230455 DOI: 10.1016/j.neulet.2023.137309] [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: 03/10/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023]
Abstract
AIMS This study examines the impact of maternal diabetes on the expression of GABAB1, GABAAα1, and mGlu2 receptors in the primary visual cortex layers of male rat newborns. MAIN METHODS In diabetic group (Dia), diabetes was induced in adult female rats using an intraperitoneal dose of Streptozotocin (STZ) 65 (mg/kg). Diabetes was managed by daily subcutaneous injection of NPH insulin in insulin-treated diabetic group (Ins). Control group (Con) received normal saline intraperitoneally rather than STZ. Male offspring born to each group of female rats were euthanized via CO2 inhalation at P0, P7, and P14 days after delivery and the expression of GABAB1, GABAAα1, and mGlu2 receptors in their primary visual cortex was determined using immunohistochemistry (IHC). KEY FINDINGS The expression of GABAB1, GABAAα1, and mGlu2 receptors increased gradually with age in the male offspring born to Con group while the highest expression was detected in layer IV of the primary visual cortex. In Dia group newborns, the expression of these receptors was significantly reduced in all layers of the primary visual cortex at every three days. Insulin treatment in diabetic mothers restored the expression of these receptors to normal levels in their newborns. SIGNIFICANCE The study indicates that diabetes reduces the expression of GABAB1, GABAAα1, and mGlu2 receptors in the primary visual cortex of male offspring born to diabetic rats at P0, P7, and P14. However, insulin treatment can counteract these effects.
Collapse
Affiliation(s)
- Javad Bagheri
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Somaye Fallahnezhad
- Nervous System Stem Cell Research Center, Semnan University of Medical Sciences, Semnan, Iran; Department of Anatomical Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Nasim Alipour
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hamideh Babaloo
- Regenerative Medicine, Organ Procurement and Transplantation Multidisciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Tahmasebi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hamed Kheradmand
- Department of Neurosurgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ghasem Sazegar
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hossein Haghir
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetic Research Center (MGRC), School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Żakowski W, Zawistowski P. Neurochemistry of the mammillary body. Brain Struct Funct 2023; 228:1379-1398. [PMID: 37378855 PMCID: PMC10335970 DOI: 10.1007/s00429-023-02673-4] [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: 10/25/2022] [Accepted: 06/19/2023] [Indexed: 06/29/2023]
Abstract
The mammillary body (MB) is a component of the extended hippocampal system and many studies have shown that its functions are vital for mnemonic processes. Together with other subcortical structures, such as the anterior thalamic nuclei and tegmental nuclei of Gudden, the MB plays a crucial role in the processing of spatial and working memory, as well as navigation in rats. The aim of this paper is to review the distribution of various substances in the MB of the rat, with a description of their possible physiological roles. The following groups of substances are reviewed: (1) classical neurotransmitters (glutamate and other excitatory transmitters, gamma-aminobutyric acid, acetylcholine, serotonin, and dopamine), (2) neuropeptides (enkephalins, substance P, cocaine- and amphetamine-regulated transcript, neurotensin, neuropeptide Y, somatostatin, orexins, and galanin), and (3) other substances (calcium-binding proteins and calcium sensor proteins). This detailed description of the chemical parcellation may facilitate a better understanding of the MB functions and its complex relations with other structures of the extended hippocampal system.
Collapse
Affiliation(s)
- Witold Żakowski
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland.
| | - Piotr Zawistowski
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| |
Collapse
|
4
|
Mozafari R, Karimi-Haghighi S, Fattahi M, Kalivas P, Haghparast A. A review on the role of metabotropic glutamate receptors in neuroplasticity following psychostimulant use disorder. Prog Neuropsychopharmacol Biol Psychiatry 2023; 124:110735. [PMID: 36813105 DOI: 10.1016/j.pnpbp.2023.110735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/03/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023]
Abstract
Psychostimulant Use Disorder (PUD) is a chronic relapsing disorder with high motivation for drug abuse. In addition to developing PUD, the use of psychostimulants is a growing public health concern because it is associated with several physical and mental health impairments. To date, there are no FDA-confirmed medicines for the treatment of psychostimulant abuse; therefore, clarification of the cellular and molecular alterations participating in PUD is crucial for developing beneficial medications. PUD causes extensive neuroadaptations in glutamatergic circuitry involved in reinforcement and reward processing. These adaptations include both transient and long-lasting changes in glutamate transmission and glutamate receptors, especially metabotropic glutamate receptors, that have been linked to developing and maintaining PUD. Here, we review the roles of all groups of mGluRs,including I,II, and III in synaptic plasticity within brain reward circuitry engaged by psychostimulants (cocaine, amphetamine, methamphetamine, and nicotine). The review concentrates on investigations of psychostimulant-induced behavioral and neurological plasticity, with an ultimate goal to explore circuit and molecular targets with the potential to contribute to the treatment of PUD.
Collapse
Affiliation(s)
- Roghayeh Mozafari
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeideh Karimi-Haghighi
- Community Based Psychiatric Care Research Center, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojdeh Fattahi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peter Kalivas
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Mazzitelli M, Presto P, Antenucci N, Meltan S, Neugebauer V. Recent Advances in the Modulation of Pain by the Metabotropic Glutamate Receptors. Cells 2022; 11:2608. [PMID: 36010684 PMCID: PMC9406805 DOI: 10.3390/cells11162608] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 01/22/2023] Open
Abstract
Metabotropic glutamate receptors (mGluR or mGlu) are G-protein coupled receptors activated by the binding of glutamate, the main classical neurotransmitter of the nervous system. Eight different mGluR subtypes (mGluR1-8) have been cloned and are classified in three groups based on their molecular, pharmacological and signaling properties. mGluRs mediate several physiological functions such as neuronal excitability and synaptic plasticity, but they have also been implicated in numerous pathological conditions including pain. The availability of new and more selective allosteric modulators together with the canonical orthosteric ligands and transgenic technologies has led to significant advances in our knowledge about the role of the specific mGluR subtypes in the pathophysiological mechanisms of various diseases. Although development of successful compounds acting on mGluRs for clinical use has been scarce, the subtype-specific-pharmacological manipulation might be a compelling approach for the treatment of several disorders in humans, including pain; this review aims to summarize and update on preclinical evidence for the roles of different mGluRs in the pain system and discusses knowledge gaps regarding mGluR-related sex differences and neuroimmune signaling in pain.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Nico Antenucci
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Shakira Meltan
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
6
|
Yuan G, Dhaynaut M, Guehl NJ, Afshar S, Huynh D, Moon SH, Iyengar SM, Jain MK, Pickett JE, Kang HJ, Ondrechen MJ, El Fakhri G, Normandin MD, Brownell AL. Design, Synthesis, and Characterization of [ 18F]mG2P026 as a High-Contrast PET Imaging Ligand for Metabotropic Glutamate Receptor 2. J Med Chem 2022; 65:9939-9954. [PMID: 35802702 PMCID: PMC9434700 DOI: 10.1021/acs.jmedchem.2c00593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An array of triazolopyridines based on JNJ-46356479 (6) were synthesized as potential positron emission tomography radiotracers for metabotropic glutamate receptor 2 (mGluR2). The selected candidates 8-10 featured enhanced positive allosteric modulator (PAM) activity (20-fold max.) and mGluR2 agonist activity (25-fold max.) compared to compound 6 in the cAMP GloSensor assays. Radiolabeling of compounds 8 and 9 (mG2P026) was achieved via Cu-mediated radiofluorination with satisfactory radiochemical yield, >5% (non-decay-corrected); high molar activity, >180 GBq/μmol; and excellent radiochemical purity, >98%. Preliminary characterization of [18F]8 and [18F]9 in rats confirmed their excellent brain permeability and binding kinetics. Further evaluation of [18F]9 in a non-human primate confirmed its superior brain heterogeneity in mapping mGluR2 and higher affinity than [18F]6. Pretreatment with different classes of PAMs in rats and a primate led to similarly enhanced brain uptake of [18F]9. As a selective ligand, [18F]9 has the potential to be developed for translational studies.
Collapse
Affiliation(s)
- Gengyang Yuan
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Nicolas J Guehl
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Sepideh Afshar
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Dalena Huynh
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Sung-Hyun Moon
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Suhasini M Iyengar
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Manish Kumar Jain
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Julie E Pickett
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Hye Jin Kang
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Mary Jo Ondrechen
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
7
|
Garceau C, Samaha AN, Cordahi T, Servonnet A, Khoo SYS. Metabotropic group II glutamate receptors in the basolateral amygdala mediate cue-triggered increases in incentive motivation. Psychopharmacology (Berl) 2021; 238:2905-2917. [PMID: 34223950 DOI: 10.1007/s00213-021-05907-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/14/2021] [Indexed: 10/20/2022]
Abstract
RATIONALE Reward-associated cues can trigger incentive motivation for reward and invigorate reward-seeking behaviour via Pavlovian-to-instrumental transfer (PIT). Glutamate signaling within the basolateral amygdala (BLA) modulates cue-triggered increases in incentive motivation. However, the role of BLA metabotropic group II glutamate (mGlu2/3) receptors is largely unknown. OBJECTIVES In Experiment 1, we characterized cue-triggered increases in incentive motivation for water reward using the PIT paradigm. In Experiment 2, we assessed the influence of intra-BLA microinjections of the mGlu2/3 receptor agonist LY379268 on this effect. METHODS Water-restricted male Sprague-Dawley rats learned to press a lever for water. Separately, they learned to associate one of two auditory cues with free water. On test days, rats could lever press under extinction conditions (no water), with intermittent, non-contingent CS+ and CS- presentations. In Experiment 1, rats were tested under baseline conditions. In Experiment 2, rats received intra-BLA microinjections of LY379268 (0, 3 and 6 [Formula: see text]g/hemisphere) before testing. RESULTS Across experiments, CS+, but not CS-, presentations increased water-associated lever pressing during testing, even though responding was reinforced neither by water nor the CS+. Intra-BLA LY379268 abolished both CS+ potentiated pressing on the water-associated lever and CS+ evoked conditioned approach to the site of water delivery. LY379268 did not influence locomotion or instrumental and Pavlovian response rates during intervals between CS presentations or during the CS-, indicating no motor effects. CONCLUSIONS mGlu2/3 receptor activity in the BLA mediates cue-triggered potentiation of incentive motivation for reward, suppressing both cue-induced increases in instrumental pursuit of the reward and anticipatory approach behaviour.
Collapse
Affiliation(s)
- Caroline Garceau
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Anne-Noël Samaha
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada. .,CNS Research Group, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
| | - Thomas Cordahi
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Alice Servonnet
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Shaun Yon-Seng Khoo
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| |
Collapse
|
8
|
Xiang Z, Lv X, Lin X, O'Brien DE, Altman MK, Lindsley CW, Javitch JA, Niswender CM, Conn PJ. Input-specific regulation of glutamatergic synaptic transmission in the medial prefrontal cortex by mGlu 2/mGlu 4 receptor heterodimers. Sci Signal 2021; 14:14/677/eabd2319. [PMID: 33824180 DOI: 10.1126/scisignal.abd2319] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are G protein-coupled receptors that regulate various aspects of central nervous system processing in normal physiology and in disease. They are thought to function as disulfide-linked homodimers, but studies have suggested that mGluRs can form functional heterodimers in cell lines. Using selective allosteric ligands, ex vivo brain slice electrophysiology, and optogenetic approaches, we found that two mGluR subtypes-mGluR2 and mGluR4 (or mGlu2 and mGlu4)-exist as functional heterodimers that regulate excitatory transmission in a synapse-specific manner within the rodent medial prefrontal cortex (mPFC). Activation of mGlu2/mGlu4 heterodimers inhibited glutamatergic signaling at thalamo-mPFC synapses but not at hippocampus-mPFC or amygdala-mPFC synapses. These findings raise the possibility that selectively targeting these heterodimers could be a therapeutic strategy for some neurologic and neuropsychiatric disorders involving specific brain circuits.
Collapse
Affiliation(s)
- Zixiu Xiang
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Xiaohui Lv
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Xin Lin
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Daniel E O'Brien
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Molly K Altman
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA.,Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY 10032, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA. .,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
9
|
Chen S, Kadakia F, Davidson S. Group II metabotropic glutamate receptor expressing neurons in anterior cingulate cortex become sensitized after inflammatory and neuropathic pain. Mol Pain 2021; 16:1744806920915339. [PMID: 32326814 PMCID: PMC7227149 DOI: 10.1177/1744806920915339] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The anterior cingulate cortex is a limbic region associated with the emotional processing of pain. How neuropathic and inflammatory pain models alter the neurophysiology of specific subsets of neurons in the anterior cingulate cortex remains incompletely understood. Here, we used a GRM2Cre:tdtomato reporter mouse line to identify a population of pyramidal neurons selectively localized to layer II/III of the murine anterior cingulate cortex. GRM2encodes the group II metabotropic glutamate receptor subtype 2 which possesses analgesic properties in mouse and human models, although its function in the anterior cingulate cortex is not known. The majority of GRM2-tdtomato anterior cingulate cortex neurons expressed GRM2gene product in situ but did not overlap with cortical markers of local inhibitory interneurons, parvalbumin or somatostatin. Physiological properties of GRM2-tdtomato anterior cingulate cortex neurons were investigated using whole-cell patch clamp techniques in slice from animals with neuropathic or inflammatory pain, and controls. After hind-paw injection of Complete Freund’s Adjuvant or chronic constriction injury, GRM2-tdtomato anterior cingulate cortex neurons exhibited enhanced excitability as measured by an increase in the number of evoked action potentials and a decreased rheobase. This hyperexcitability was reversed pharmacologically by bath application of the metabotropic glutamate receptor subtype 2 agonist (2R, 4R)-4-Aminopyrrolidine-2,4-dicarboxylate APDC (1 µM) in both inflammatory and neuropathic models. We conclude that layer II/III pyramidal GRM2-tdtomato anterior cingulate cortex neurons express functional group II metabotropic glutamate receptors and undergo changes to membrane biophysical properties under conditions of inflammatory and neuropathic pain.
Collapse
Affiliation(s)
- Sisi Chen
- Department of Anesthesiology, Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Feni Kadakia
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Steve Davidson
- Department of Anesthesiology, Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
10
|
Steullet P. Thalamus-related anomalies as candidate mechanism-based biomarkers for psychosis. Schizophr Res 2020; 226:147-157. [PMID: 31147286 DOI: 10.1016/j.schres.2019.05.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 02/08/2023]
Abstract
Identification of reliable biomarkers of prognosis in subjects with high risk to psychosis is an essential step to improve care and treatment of this population of help-seekers. Longitudinal studies highlight some clinical criteria, cognitive deficits, patterns of gray matter alterations and profiles of blood metabolites that provide some levels of prediction regarding the conversion to psychosis. Further effort is warranted to validate these results and implement these types of approaches in clinical settings. Such biomarkers may however fall short in entangling the biological mechanisms underlying the disease progression, an essential step in the development of novel therapies. Circuit-based approaches, which map on well-identified cerebral functions, could meet these needs. Converging evidence indicates that thalamus abnormalities are central to schizophrenia pathophysiology, contributing to clinical symptoms, cognitive and sensory deficits. This review highlights the various thalamus-related anomalies reported in individuals with genetic risks and in the different phases of the disorder, from prodromal to chronic stages. Several anomalies are potent endophenotypes, while others exist in clinical high-risk subjects and worsen in those who convert to full psychosis. Aberrant functional coupling between thalamus and cortex, low glutamate content and readouts from resting EEG carry predictive values for transition to psychosis or functional outcome. In this context, thalamus-related anomalies represent a valuable entry point to tackle circuit-based alterations associated with the emergence of psychosis. This review also proposes that longitudinal surveys of neuroimaging, EEG readouts associated with circuits encompassing the mediodorsal, pulvinar in high-risk individuals could unveil biological mechanisms contributing to this psychiatric disorder.
Collapse
Affiliation(s)
- Pascal Steullet
- Center of Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, Site de Cery, 1008 Prilly-Lausanne, Switzerland.
| |
Collapse
|
11
|
Yuan G, Qu X, Zheng B, Neelamegam R, Afshar S, Iyengar S, Pan C, Wang J, Kang HJ, Ondrechen MJ, Poutiainen P, El Fakhri G, Zhang Z, Brownell AL. Design, Synthesis, and Characterization of Benzimidazole Derivatives as Positron Emission Tomography Imaging Ligands for Metabotropic Glutamate Receptor 2. J Med Chem 2020; 63:12060-12072. [PMID: 32981322 DOI: 10.1021/acs.jmedchem.0c01394] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Three benzimidazole derivatives (13-15) have been synthetized as potential positron emission tomography (PET) imaging ligands for mGluR2 in the brain. Of these compounds, 13 exhibits potent binding affinity (IC50 = 7.6 ± 0.9 nM), positive allosteric modulator (PAM) activity (EC50 = 51.2 nM), and excellent selectivity against other mGluR subtypes (>100-fold). [11C]13 was synthesized via O-[11C]methylation of its phenol precursor 25 with [11C]methyl iodide. The achieved radiochemical yield was 20 ± 2% (n = 10, decay-corrected) based on [11C]CO2 with a radiochemical purity of >98% and molar activity of 98 ± 30 GBq/μmol EOS. Ex vivo biodistribution studies revealed reversible accumulation of [11C]13 and hepatobiliary and urinary excretions. PET imaging studies in rats demonstrated that [11C]13 accumulated in the mGluR2-rich brain regions. Pre-administration of mGluR2-selective PAM, 17 reduced the brain uptake of [11C]13, indicating a selective binding. Therefore, [11C]13 is a potential PET imaging ligand for mGluR2 in different central nervous system-related conditions.
Collapse
Affiliation(s)
- Gengyang Yuan
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Xiying Qu
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Baohui Zheng
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Sepideh Afshar
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Suhasini Iyengar
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Chuzhi Pan
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Junfeng Wang
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Hye Jin Kang
- Department of Pharmacology, University of North Carolina Chapel Hill School of Medicine, Chapel Hill, North Carolina 27514, United States
| | - Mary Jo Ondrechen
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Pekka Poutiainen
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio 70210, Finland
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Zhaoda Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Suite 2301, Charlestown, Massachusetts 02129, United States
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
12
|
Molosh AI, Dustrude ET, Lukkes JL, Fitz SD, Caliman IF, Abreu ARR, Dietrich AD, Truitt WA, Ver Donck L, Ceusters M, Kent JM, Johnson PL, Shekhar A. Panic results in unique molecular and network changes in the amygdala that facilitate fear responses. Mol Psychiatry 2020; 25:442-460. [PMID: 30108314 PMCID: PMC6410355 DOI: 10.1038/s41380-018-0119-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 04/03/2018] [Accepted: 05/25/2018] [Indexed: 11/12/2022]
Abstract
Recurrent panic attacks (PAs) are a common feature of panic disorder (PD) and post-traumatic stress disorder (PTSD). Several distinct brain regions are involved in the regulation of panic responses, such as perifornical hypothalamus (PeF), periaqueductal gray, amygdala and frontal cortex. We have previously shown that inhibition of GABA synthesis in the PeF produces panic-vulnerable rats. Here, we investigate the mechanisms by which a panic-vulnerable state could lead to persistent fear. We first show that optogenetic activation of glutamatergic terminals from the PeF to the basolateral amygdala (BLA) enhanced the acquisition, delayed the extinction and induced the persistence of fear responses 3 weeks later, confirming a functional PeF-amygdala pathway involved in fear learning. Similar to optogenetic activation of PeF, panic-prone rats also exhibited delayed extinction. Next, we demonstrate that panic-prone rats had altered inhibitory and enhanced excitatory synaptic transmission of the principal neurons, and reduced protein levels of metabotropic glutamate type 2 receptor (mGluR2) in the BLA. Application of an mGluR2-positive allosteric modulator (PAM) reduced glutamate neurotransmission in the BLA slices from panic-prone rats. Treating panic-prone rats with mGluR2 PAM blocked sodium lactate (NaLac)-induced panic responses and normalized fear extinction deficits. Finally, in a subset of patients with comorbid PD, treatment with mGluR2 PAM resulted in complete remission of panic symptoms. These data demonstrate that a panic-prone state leads to specific reduction in mGluR2 function within the amygdala network and facilitates fear, and mGluR2 PAMs could be a targeted treatment for panic symptoms in PD and PTSD patients.
Collapse
Affiliation(s)
- A I Molosh
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Paul and Carol Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E T Dustrude
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J L Lukkes
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S D Fitz
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - I F Caliman
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A R R Abreu
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A D Dietrich
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - W A Truitt
- Paul and Carol Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - L Ver Donck
- Janssen Research & Development, Beerse, Belgium
| | - M Ceusters
- Janssen Research & Development, Beerse, Belgium
| | - J M Kent
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | - P L Johnson
- Paul and Carol Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Shekhar
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
- Paul and Carol Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Clinical and Translational Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
13
|
Branched Photoswitchable Tethered Ligands Enable Ultra-efficient Optical Control and Detection of G Protein-Coupled Receptors In Vivo. Neuron 2019; 105:446-463.e13. [PMID: 31784287 DOI: 10.1016/j.neuron.2019.10.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/26/2019] [Accepted: 10/27/2019] [Indexed: 01/01/2023]
Abstract
The limitations of classical drugs have spurred the development of covalently tethered photoswitchable ligands to control neuromodulatory receptors. However, a major shortcoming of tethered photopharmacology is the inability to obtain optical control with an efficacy comparable with that of the native ligand. To overcome this, we developed a family of branched photoswitchable compounds to target metabotropic glutamate receptors (mGluRs). These compounds permit photo-agonism of Gi/o-coupled group II mGluRs with near-complete efficiency relative to glutamate when attached to receptors via a range of orthogonal, multiplexable modalities. Through a chimeric approach, branched ligands also allow efficient optical control of Gq-coupled mGluR5, which we use to probe the spatiotemporal properties of receptor-induced calcium oscillations. In addition, we report branched, photoswitch-fluorophore compounds for simultaneous receptor imaging and manipulation. Finally, we demonstrate this approach in vivo in mice, where photoactivation of SNAP-mGluR2 in the medial prefrontal cortex reversibly modulates working memory in normal and disease-associated states.
Collapse
|
14
|
Mazzitelli M, Neugebauer V. Amygdala group II mGluRs mediate the inhibitory effects of systemic group II mGluR activation on behavior and spinal neurons in a rat model of arthritis pain. Neuropharmacology 2019; 158:107706. [PMID: 31306647 DOI: 10.1016/j.neuropharm.2019.107706] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022]
Abstract
The amygdala plays a critical role in emotional-affective aspects of behaviors and pain modulation. The central nucleus of amygdala (CeA) serves major output functions, and neuroplasticity in the CeA is linked to pain-related behaviors in different models. Activation of Gi/o-coupled group II metabotropic glutamate receptors (mGluRs), which consist of mGluR2 and mGluR3, can decrease neurotransmitter release and regulate synaptic plasticity. Group II mGluRs have emerged as targets for neuropsychiatric disorders and can inhibit pain-related processing and behaviors. Surprisingly, site and mechanism of antinociceptive actions of systemically applied group II mGluR agonists are not clear. Our previous work showed that group II mGluR activation in the amygdala inhibits pain-related CeA activity, but behavioral and spinal consequences remain to be determined. Here we studied the contribution of group II mGluRs in the amygdala to the antinociceptive effects of a systemically applied group II mGluR agonist (LY379268) on behavior and spinal dorsal horn neuronal activity, using the kaolin/carrageenan-induced knee joint arthritis pain model. Audible and ultrasonic vocalizations (emotional responses) and mechanical reflex thresholds were measured in adult rats with and without arthritis (5-6 h postinduction). Extracellular single-unit recordings were made from spinal dorsal horn wide dynamic range neurons of anesthetized (isoflurane) rats with and without arthritis (5-6 h postinduction). Systemic (intraperitoneal) application of a group II mGluR agonist (LY379268) decreased behaviors and activity of spinal neurons in the arthritis pain model but not under normal conditions. Stereotaxic administration of LY379268 into the CeA mimicked the effects of systemic application. Conversely, stereotaxic administration of a group II mGluR antagonist (LY341495) into the CeA reversed the effects of systemic application of LY379268 on behaviors and dorsal horn neuronal activity in arthritic rats. The data show for the first time that the amygdala is the critical site of action for the antinociceptive behavioral and spinal neuronal effects of systemically applied group II mGluR agonists.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA; Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA.
| |
Collapse
|
15
|
Abstract
For decades, symptoms of depression have been treated primarily with medications that directly target the monoaminergic brain systems, which typically take weeks to exert measurable effects and months to exert remission of symptoms. Low, subanesthetic doses of ( R,S)-ketamine (ketamine) result in the rapid improvement of core depressive symptoms, including mood, anhedonia, and suicidal ideation, occurring within hours following a single administration, with relief from symptoms typically lasting up to a week. The discovery of these actions of ketamine has resulted in a reconceptualization of how depression could be more effectively treated in the future. In this review, we discuss clinical data pertaining to ketamine and other rapid-acting antidepressant drugs, as well as the current state of pharmacological knowledge regarding their mechanism of action. Additionally, we discuss the neurobiological circuits that are engaged by this drug class and that may be targeted by a future generation of medications, for example, hydroxynorketamine; metabotropic glutamate receptor 2/3 antagonists; and N-methyl-d-aspartate, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and γ-aminobutyric acid receptor modulators.
Collapse
Affiliation(s)
- Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA;
- Departments of Pharmacology and Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA 20892
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA;
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| |
Collapse
|
16
|
Mazzitelli M, Palazzo E, Maione S, Neugebauer V. Group II Metabotropic Glutamate Receptors: Role in Pain Mechanisms and Pain Modulation. Front Mol Neurosci 2018; 11:383. [PMID: 30356691 PMCID: PMC6189308 DOI: 10.3389/fnmol.2018.00383] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Glutamate is the main excitatory neurotransmitter in the nervous system and plays a critical role in nociceptive processing and pain modulation. G-protein coupled metabotropic glutamate receptors (mGluRs) are widely expressed in the central and peripheral nervous system, and they mediate neuronal excitability and synaptic transmission. Eight different mGluR subtypes have been identified so far, and are classified into Groups I-III. Group II mGluR2 and mGluR3 couple negatively to adenylyl cyclase through Gi/Go proteins, are mainly expressed presynaptically, and typically inhibit the release of neurotransmitters, including glutamate and GABA. Group II mGluRs have consistently been linked to pain modulation; they are expressed in peripheral, spinal and supraspinal elements of pain-related neural processing. Pharmacological studies have shown anti-nociceptive/analgesic effects of group II mGluR agonists in preclinical models of acute and chronic pain, although much less is known about mechanisms and sites of action for mGluR2 and mGluR3 compared to other mGluRs. The availability of orthosteric and new selective allosteric modulators acting on mGluR2 and mGluR3 has provided valuable tools for elucidating (subtype) specific contributions of these receptors to the pathophysiological mechanisms of pain and other disorders and their potential as therapeutic targets. This review focuses on the important role of group II mGluRs in the neurobiology of pain mechanisms and behavioral modulation, and discusses evidence for their therapeutic potential in pain.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Enza Palazzo
- Section of Pharmacology L. Donatelli, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Section of Pharmacology L. Donatelli, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
17
|
Cross AJ, Anthenelli R, Li X. Metabotropic Glutamate Receptors 2 and 3 as Targets for Treating Nicotine Addiction. Biol Psychiatry 2018; 83:947-954. [PMID: 29301614 PMCID: PMC5953779 DOI: 10.1016/j.biopsych.2017.11.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 11/02/2017] [Accepted: 11/14/2017] [Indexed: 12/27/2022]
Abstract
Tobacco smoking, driven by the addictive properties of nicotine, continues to be a worldwide health problem. Based on the well-established role of glutamatergic neurotransmission in drug addiction, novel medication development strategies seek to halt nicotine consumption and prevent relapse to tobacco smoking by modulating glutamate transmission. The presynaptic inhibitory metabotropic glutamate receptors 2 and 3 (mGluR2/3) are key autoreceptors on glutamatergic terminals that maintain glutamate homeostasis. Accumulating evidence suggests the critical role of mGluR2/3 in different aspects of nicotine addiction, including acquisition and maintenance of nicotine taking, nicotine withdrawal, and persistent nicotine seeking even after prolonged abstinence. The involvement of mGluR2/3 in other neuropsychiatric conditions, such as anxiety, depression, schizophrenia, Alzheimer's disease, Parkinson's disease, and pain, provides convincing evidence suggesting that mGluR2/3 may provide an effective therapeutic approach for comorbidity of smoking and these conditions. This focused review article highlights that mGluR2/3 provide a promising target in the search for smoking cessation medication with novel mechanisms of actions that differ from those of currently U.S. Food and Drug Administration-approved pharmacotherapies.
Collapse
Affiliation(s)
- Alan J Cross
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, Massachusetts
| | - Robert Anthenelli
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, California
| | - Xia Li
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
18
|
Gupta I, Young AMJ. Metabotropic glutamate receptor modulation of dopamine release in the nucleus accumbens shell is unaffected by phencyclidine pretreatment: In vitro assessment using fast-scan cyclic voltammetry rat brain slices. Brain Res 2018. [PMID: 29524437 DOI: 10.1016/j.brainres.2018.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The non-competitive glutamate antagonist, phencyclidine is used in rodents to model behavioural deficits see in schizophrenia. Importantly, these deficits endure long after the cessation of short-term chronic treatment (sub-chronic), indicating that the drug treatment causes long-term changes in the physiology and/or chemistry of the brain. There is evidence that this may occur through glutamatergic modulation of mesolimbic dopamine release, perhaps involving metabotropic glutamate receptors (mGluR). This study sought to investigate the effect of sub-chronic phencyclidine pretreatment on modulation of dopamine neurotransmission by metabotropic glutamate receptors 2 and 5 (mGluR2 and mGluR5) in the nucleus accumbens shell in vitro, with the hypothesis that phencyclidine pretreatment would disrupt the mGluR-mediated modulation of dopamine release. We showed that the orthosteric mGluR2 agonist LY379268 (0.1 µM, 1 µM and 10 µM) and mGluR5 positive allosteric modulator CDPPB (1 µM and 10 µM) both attenuated potassium-evoked dopamine release, underscoring their role in modulating dopamine neurotransmission in the nucleus accumbens. Sub-chronic PCP treatment, which caused cognitive deficits measured by performance in the novel object recognition task, modelling aspects of behavioral deficits seen in schizophrenia, induced neurobiological changes that enhanced dopamine release in the nucleus accumbens, but had no effect on mGluR2 or mGluR5 mediated changes in dopamine release. Therefore it is unlikely that schizophrenia-related behavioural changes seen after sub-chronic phencyclidine pre-treatment are mediated through mGluR modulation of dopamine release.
Collapse
Affiliation(s)
- Ishan Gupta
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Andrew M J Young
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK.
| |
Collapse
|
19
|
Monn JA, Henry SS, Massey SM, Clawson DK, Chen Q, Diseroad BA, Bhardwaj RM, Atwell S, Lu F, Wang J, Russell M, Heinz BA, Wang XS, Carter JH, Getman BG, Adragni K, Broad LM, Sanger HE, Ursu D, Catlow JT, Swanson S, Johnson BG, Shaw DB, McKinzie DL, Hao J. Synthesis and Pharmacological Characterization of C4 β-Amide-Substituted 2-Aminobicyclo[3.1.0]hexane-2,6-dicarboxylates. Identification of (1 S,2 S,4 S,5 R,6 S)-2-Amino-4-[(3-methoxybenzoyl)amino]bicyclo[3.1.0]hexane-2,6-dicarboxylic Acid (LY2794193), a Highly Potent and Selective mGlu 3 Receptor Agonist. J Med Chem 2018; 61:2303-2328. [PMID: 29350927 DOI: 10.1021/acs.jmedchem.7b01481] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Multiple therapeutic opportunities have been suggested for compounds capable of selective activation of metabotropic glutamate 3 (mGlu3) receptors, but small molecule tools are lacking. As part of our ongoing efforts to identify potent, selective, and systemically bioavailable agonists for mGlu2 and mGlu3 receptor subtypes, a series of C4β-N-linked variants of (1 S,2 S,5 R,6 S)-2-amino-bicyclo[3.1.0]hexane-2,6-dicarboxylic acid 1 (LY354740) were prepared and evaluated for both mGlu2 and mGlu3 receptor binding affinity and functional cellular responses. From this investigation we identified (1 S,2 S,4 S,5 R,6 S)-2-amino-4-[(3-methoxybenzoyl)amino]bicyclo[3.1.0]hexane-2,6-dicarboxylic acid 8p (LY2794193), a molecule that demonstrates remarkable mGlu3 receptor selectivity. Crystallization of 8p with the amino terminal domain of hmGlu3 revealed critical binding interactions for this ligand with residues adjacent to the glutamate binding site, while pharmacokinetic assessment of 8p combined with its effect in an mGlu2 receptor-dependent behavioral model provides estimates for doses of this compound that would be expected to selectively engage and activate central mGlu3 receptors in vivo.
Collapse
|
20
|
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: 2.4] [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
|
21
|
Li DP, Pan HL. Glutamatergic Regulation of Hypothalamic Presympathetic Neurons in Hypertension. Curr Hypertens Rep 2017; 19:78. [PMID: 28929331 DOI: 10.1007/s11906-017-0776-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Elevated sympathetic vasomotor tone emanating from the brain is a major mechanism involved in the development of hypertension. Increased glutamatergic excitatory input to presympathetic neurons in the paraventricular nucleus (PVN) of the hypothalamus leads to increased sympathetic outflow in various animal models of hypertension. Recent studies have revealed molecular and cellular mechanisms underlying enhanced glutamatergic synaptic input to PVN presympathetic neurons in hypertension. In this review article, we summarize recent findings on changes in inotropic and metabotropic glutamate receptors, at both presynaptic and postsynaptic sites, responsible for increased glutamatergic input to PVN presympathetic neurons in hypertension. Particular emphasis is placed on the role of protein kinases and phosphatases in the potentiated activity of synaptic NMDA receptors in the PVN in hypertension. New findings about glutamatergic synaptic plasticity in the PVN not only improve the understanding of molecular mechanisms involved in heightened activity of the sympathetic nervous system but also suggest new therapeutic targets for treating drug-resistant, neurogenic hypertension.
Collapse
Affiliation(s)
- De-Pei Li
- Center for Neuroscience and Pain Research, Division of Anesthesiology and Critical Care, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Division of Anesthesiology and Critical Care, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
22
|
|
23
|
Moreno Delgado D, Møller TC, Ster J, Giraldo J, Maurel D, Rovira X, Scholler P, Zwier JM, Perroy J, Durroux T, Trinquet E, Prezeau L, Rondard P, Pin JP. Pharmacological evidence for a metabotropic glutamate receptor heterodimer in neuronal cells. eLife 2017; 6. [PMID: 28661401 PMCID: PMC5540479 DOI: 10.7554/elife.25233] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 06/26/2017] [Indexed: 12/25/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs) are mandatory dimers playing important roles in regulating CNS function. Although assumed to form exclusive homodimers, 16 possible heterodimeric mGluRs have been proposed but their existence in native cells remains elusive. Here, we set up two assays to specifically identify the pharmacological properties of rat mGlu heterodimers composed of mGlu2 and 4 subunits. We used either a heterodimer-specific conformational LRET-based biosensor or a system that guarantees the cell surface targeting of the heterodimer only. We identified mGlu2-4 specific pharmacological fingerprints that were also observed in a neuronal cell line and in lateral perforant path terminals naturally expressing mGlu2 and mGlu4. These results bring strong evidence for the existence of mGlu2-4 heterodimers in native cells. In addition to reporting a general approach to characterize heterodimeric mGluRs, our study opens new avenues to understanding the pathophysiological roles of mGlu heterodimers. DOI:http://dx.doi.org/10.7554/eLife.25233.001
Collapse
Affiliation(s)
- David Moreno Delgado
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Thor C Møller
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Jeanne Ster
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Network Biomedical Research Center on Mental Health, Madrid, Spain
| | - Damien Maurel
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Xavier Rovira
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Pauline Scholler
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | | | - Julie Perroy
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | | | - Laurent Prezeau
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Philippe Rondard
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle (IGF), CNRS, INSERM, Univ. Montpellier, Montpellier, France
| |
Collapse
|
24
|
Johnson MP, Muhlhauser MA, Nisenbaum ES, Simmons RMA, Forster BM, Knopp KL, Yang L, Morrow D, Li DL, Kennedy JD, Swanson S, Monn JA. Broad spectrum efficacy with LY2969822, an oral prodrug of metabotropic glutamate 2/3 receptor agonist LY2934747, in rodent pain models. Br J Pharmacol 2017; 174:822-835. [PMID: 28177520 DOI: 10.1111/bph.13740] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 01/27/2017] [Accepted: 01/31/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE A body of evidence suggests activation of metabotropic glutamate 2/3 (mGlu2/3 ) receptors would be an effective analgesic in chronic pain conditions. Thus, the analgesic properties of a novel mGlu2/3 receptor agonist prodrug were investigated. EXPERIMENTAL APPROACH After oral absorption, the prodrug LY2969822 rapidly converts to the brain penetrant, potent and subtype-selective mGlu2/3 receptor agonist LY2934747. Behavioural assessments of allodynia, hyperalgesia and nocifensive behaviours were determined in preclinical pain models after administration of LY2969822 0.3-10 mg·kg-1 . In addition, the ability of i.v. LY2934747 to modulate dorsal horn spinal cord wide dynamic range (WDR) neurons in spinal nerve ligated (SNL) rats was assessed. KEY RESULTS Following treatment with LY2934747, the spontaneous activity and electrically-evoked wind-up of WDR neurons in rats that had undergone spinal nerve ligation and developed mechanical allodynia were suppressed. In a model of sensitization, orally administered LY2969822 prevented the nociceptive behaviours induced by an intraplantar injection of formalin. The on-target nature of this effect was confirmed by blockade with an mGlu2/3 receptor antagonist. LY2969822 prevented capsaicin-induced tactile hypersensitivity, reversed the SNL-induced tactile hypersensitivity and reversed complete Freund's adjuvant - induced mechanical hyperalgesia. The mGlu2/3 receptor agonist prodrug demonstrated efficacy in visceral pain models, including a colorectal distension model and partially prevented the nocifensive behaviours in the mouse acetic acid writhing model. CONCLUSIONS AND IMPLICATIONS Following oral administration of the prodrug LY2969822, the mGlu2/3 receptor agonist LY2934747 was formed and this attenuated pain behaviours across a broad range of preclinical pain models.
Collapse
Affiliation(s)
- Michael P Johnson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mark A Muhlhauser
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Eric S Nisenbaum
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Rosa M A Simmons
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Beth M Forster
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Kelly L Knopp
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Lijuan Yang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Denise Morrow
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Dominic L Li
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jeffrey D Kennedy
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Steven Swanson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - James A Monn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
25
|
Chappell MD, Li R, Smith SC, Dressman BA, Tromiczak EG, Tripp AE, Blanco MJ, Vetman T, Quimby SJ, Matt J, Britton TC, Fivush AM, Schkeryantz JM, Mayhugh D, Erickson JA, Bures MG, Jaramillo C, Carpintero M, Diego JED, Barberis M, Garcia-Cerrada S, Soriano JF, Antonysamy S, Atwell S, MacEwan I, Condon B, Sougias C, Wang J, Zhang A, Conners K, Groshong C, Wasserman SR, Koss JW, Witkin JM, Li X, Overshiner C, Wafford KA, Seidel W, Wang XS, Heinz BA, Swanson S, Catlow JT, Bedwell DW, Monn JA, Mitch CH, Ornstein PL. Discovery of (1S,2R,3S,4S,5R,6R)-2-Amino-3-[(3,4-difluorophenyl)sulfanylmethyl]-4-hydroxy-bicyclo[3.1.0]hexane-2,6-dicarboxylic Acid Hydrochloride (LY3020371·HCl): A Potent, Metabotropic Glutamate 2/3 Receptor Antagonist with Antidepressant-Like Activity. J Med Chem 2016; 59:10974-10993. [DOI: 10.1021/acs.jmedchem.6b01119] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Carlos Jaramillo
- Discovery
Chemistry Synthesis Group, Centro de Investigación Lilly S.A. Avda. de la Industria, 30 Alcobendas, Madrid 28108, Spain
| | - Mercedes Carpintero
- Discovery
Chemistry Synthesis Group, Centro de Investigación Lilly S.A. Avda. de la Industria, 30 Alcobendas, Madrid 28108, Spain
| | - José Eugenio de Diego
- Discovery
Chemistry Synthesis Group, Centro de Investigación Lilly S.A. Avda. de la Industria, 30 Alcobendas, Madrid 28108, Spain
| | - Mario Barberis
- Discovery
Chemistry Synthesis Group, Centro de Investigación Lilly S.A. Avda. de la Industria, 30 Alcobendas, Madrid 28108, Spain
| | - Susana Garcia-Cerrada
- Discovery
Chemistry Synthesis Group, Centro de Investigación Lilly S.A. Avda. de la Industria, 30 Alcobendas, Madrid 28108, Spain
| | - José F. Soriano
- Discovery
Chemistry Synthesis Group, Centro de Investigación Lilly S.A. Avda. de la Industria, 30 Alcobendas, Madrid 28108, Spain
| | - Stephen Antonysamy
- Structural
Biology, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Shane Atwell
- Structural
Biology, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Iain MacEwan
- Structural
Biology, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Bradley Condon
- Structural
Biology, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Christine Sougias
- Structural
Biology, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Jing Wang
- Structural
Biology, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Aiping Zhang
- Structural
Biology, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Kris Conners
- Structural
Biology, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Chris Groshong
- Structural
Biology, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Stephen R. Wasserman
- Structural Biology,
Eli Lilly and Company, Advanced Photon Source, Argonne National Laboratory, Building 438A, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | - John W. Koss
- Structural Biology,
Eli Lilly and Company, Advanced Photon Source, Argonne National Laboratory, Building 438A, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | | | | | | | - Keith A. Wafford
- Neuroscience Research, Eli Lilly and Company, Erl Wood Manor, Sunninghill Road, Windlesham, Surrey, U.K. GU20 6PH
| | - Wesley Seidel
- Neuroscience Research, Eli Lilly and Company, Erl Wood Manor, Sunninghill Road, Windlesham, Surrey, U.K. GU20 6PH
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
McOmish CE, Demireva EY, Gingrich JA. Developmental expression of mGlu2 and mGlu3 in the mouse brain. Gene Expr Patterns 2016; 22:46-53. [PMID: 27818290 DOI: 10.1016/j.gep.2016.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 10/30/2016] [Accepted: 10/31/2016] [Indexed: 12/12/2022]
Abstract
The glutamatergic system directs central nervous system (CNS) neuronal activity and may underlie various neuropsychiatric disorders. Glutamate transmits its effects through multiple receptor classes. Class II metabotropic glutamate receptors, mGlu2 and mGlu3, play an important role in regulating synaptic release of different neurotransmitter systems and consequently modulate signaling across several neuronal subtypes. Drugs targeting mGlu2 and mGlu3 are seen as potential therapeutics for various psychiatric and neurological disorders, and defining their expression through development can aid in understanding their distinct function. Here, non-radioactive in situ hybridization was used to detect mGlu2 and mGlu3 mRNA in the CNS of 129SvEv mice at PN1, PN8, PN25, PN40, and PN100. At PN1, mGlu2 and mGlu3 are strongly expressed cortically, most notably in layer III and V. Subcortically, mGlu2 is detected in thalamic nuclei; mGlu3 is highly expressed in the striatum. By PN8, the most notable changes are in hippocampus and cortex, with mGlu2 densely expressed in the dentate gyrus, and showing increased cortical levels especially in medial cortex. At PN8, mGlu3 is observed in cortex and striatum, with highest levels detected in reticular thalamic nucleus. At PN25 patterns of expression approximated those observed across adulthood (PN40 & PN100): mGlu2 expression was high in cortex and dentate gyrus while mGlu3 showed expression in the reticular thalamic nucleus, cortex, and striatum. These studies provide a foundation for future research seeking to parse out the roles of mGlu2 from mGlu3, paving the way for better understanding of how these receptors regulate activity in the brain.
Collapse
Affiliation(s)
- Caitlin E McOmish
- Molecular Psychiatry, Florey Institute for Neuroscience and Mental Health, VIC, 3052, Australia; Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Elena Y Demireva
- Michigan State University, East Lansing, MI 48824, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jay A Gingrich
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA; Sackler Institute for Developmental Psychobiology, New York, NY 10032, USA
| |
Collapse
|
27
|
Hurley MM, Maunze B, Block ME, Frenkel MM, Reilly MJ, Kim E, Chen Y, Li Y, Baker DA, Liu QS, Choi S. Pituitary Adenylate-Cyclase Activating Polypeptide Regulates Hunger- and Palatability-Induced Binge Eating. Front Neurosci 2016; 10:383. [PMID: 27597817 PMCID: PMC4993128 DOI: 10.3389/fnins.2016.00383] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/05/2016] [Indexed: 11/13/2022] Open
Abstract
While pituitary adenylate cyclase activating polypeptide (PACAP) signaling in the hypothalamic ventromedial nuclei (VMN) has been shown to regulate feeding, a challenge in unmasking a role for this peptide in obesity is that excess feeding can involve numerous mechanisms including homeostatic (hunger) and hedonic-related (palatability) drives. In these studies, we first isolated distinct feeding drives by developing a novel model of binge behavior in which homeostatic-driven feeding was temporally separated from feeding driven by food palatability. We found that stimulation of the VMN, achieved by local microinjections of AMPA, decreased standard chow consumption in food-restricted rats (e.g., homeostatic feeding); surprisingly, this manipulation failed to alter palatable food consumption in satiated rats (e.g., hedonic feeding). In contrast, inhibition of the nucleus accumbens (NAc), through local microinjections of GABA receptor agonists baclofen and muscimol, decreased hedonic feeding without altering homeostatic feeding. PACAP microinjections produced the site-specific changes in synaptic transmission needed to decrease feeding via VMN or NAc circuitry. PACAP into the NAc mimicked the actions of GABA agonists by reducing hedonic feeding without altering homeostatic feeding. In contrast, PACAP into the VMN mimicked the actions of AMPA by decreasing homeostatic feeding without affecting hedonic feeding. Slice electrophysiology recordings verified PACAP excitation of VMN neurons and inhibition of NAc neurons. These data suggest that the VMN and NAc regulate distinct circuits giving rise to unique feeding drives, but that both can be regulated by the neuropeptide PACAP to potentially curb excessive eating stemming from either drive.
Collapse
Affiliation(s)
- Matthew M Hurley
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Brian Maunze
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Megan E Block
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Mogen M Frenkel
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Michael J Reilly
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Eugene Kim
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Yao Chen
- Department of Pharmacology and Toxicology, Medical College of Wisconsin Milwaukee, WI, USA
| | - Yan Li
- Department of Pharmacology and Toxicology, Medical College of Wisconsin Milwaukee, WI, USA
| | - David A Baker
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin Milwaukee, WI, USA
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| |
Collapse
|
28
|
Muguruza C, Meana JJ, Callado LF. Group II Metabotropic Glutamate Receptors as Targets for Novel Antipsychotic Drugs. Front Pharmacol 2016; 7:130. [PMID: 27242534 PMCID: PMC4873505 DOI: 10.3389/fphar.2016.00130] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/05/2016] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a chronic psychiatric disorder which substantially impairs patients' quality of life. Despite the extensive research in this field, the pathophysiology and etiology of schizophrenia remain unknown. Different neurotransmitter systems and functional networks have been found to be affected in the brain of patients with schizophrenia. In this context, postmortem brain studies as well as genetic assays have suggested alterations in Group II metabotropic glutamate receptors (mGluRs) in schizophrenia. Despite many years of drug research, several needs in the treatment of schizophrenia have not been addressed sufficiently. In fact, only 5-10% of patients with schizophrenia successfully achieve a full recovery after treatment. In recent years mGluRs have turned up as novel targets for the design of new antipsychotic medications for schizophrenia. Concretely, Group II mGluRs are of particular interest due to their regulatory role in neurotransmission modulating glutamatergic activity in brain synapses. Preclinical studies have demonstrated that orthosteric Group II mGluR agonists exhibit antipsychotic-like properties in animal models of schizophrenia. However, when these compounds have been tested in human clinical studies with schizophrenic patients results have been inconclusive. Nevertheless, it has been recently suggested that this apparent lack of efficacy in schizophrenic patients may be related to previous exposure to atypical antipsychotics. Moreover, the role of the functional heterocomplex formed by 5-HT2A and mGlu2 receptors in the clinical response to Group II mGluR agonists is currently under study.
Collapse
Affiliation(s)
- Carolina Muguruza
- Department of Pharmacology, University of the Basque Country, UPV/EHULeioa, Spain
- Centro de Investigación Biomédica en Red de Salud MentalMadrid, Spain
| | - J. Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHULeioa, Spain
- Centro de Investigación Biomédica en Red de Salud MentalMadrid, Spain
| | - Luis F. Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHULeioa, Spain
- Centro de Investigación Biomédica en Red de Salud MentalMadrid, Spain
| |
Collapse
|
29
|
Ahnaou A, de Boer P, Lavreysen H, Huysmans H, Sinha V, Raeymaekers L, Van De Casteele T, Cid J, Van Nueten L, Macdonald G, Kemp J, Drinkenburg W. Translational neurophysiological markers for activity of the metabotropic glutamate receptor (mGluR2) modulator JNJ-40411813: Sleep EEG correlates in rodents and healthy men. Neuropharmacology 2016; 103:290-305. [DOI: 10.1016/j.neuropharm.2015.11.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/28/2015] [Accepted: 11/28/2015] [Indexed: 12/31/2022]
|
30
|
Engel M, Snikeris P, Matosin N, Newell KA, Huang XF, Frank E. mGluR2/3 agonist LY379268 rescues NMDA and GABAA receptor level deficits induced in a two-hit mouse model of schizophrenia. Psychopharmacology (Berl) 2016; 233:1349-59. [PMID: 26861891 DOI: 10.1007/s00213-016-4230-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 01/29/2016] [Indexed: 10/22/2022]
Abstract
RATIONALE An imbalance of excitatory and inhibitory neurotransmission underlies the glutamate hypothesis of schizophrenia. Agonists of group II metabotropic glutamate receptors, mGluR2/3, have been proposed as novel therapeutic agents to correct this imbalance. However, the influence of mGluR2/3 activity on excitatory and inhibitory neurotransmitter receptors has not been explored. OBJECTIVES We aimed to investigate the ability of a novel mGluR2/3 agonist, LY379268, to modulate the availability of the excitatory N-methyl-D-aspartate receptor (NMDA-R) and the inhibitory gamma-aminobutyrate-A receptor (GABAA-R), in a two-hit mouse model of schizophrenia. METHODS Wild type (WT) and heterozygous neuregulin 1 transmembrane domain mutant mice (NRG1 HET) were treated daily with phencyclidine (10 mg/kg ip) or saline for 14 days. After a 14-day washout, an acute dose of the mGluR2/3 agonist LY379268 (3 mg/kg), olanzapine (antipsychotic drug comparison, 1.5 mg/kg), or saline was administered. NMDA-R and GABAA-R binding densities were examined by receptor autoradiography in several schizophrenia-relevant brain regions. RESULTS In both WT and NRG1 HET mice, phencyclidine treatment significantly reduced NMDA-R and GABAA-R binding density in the prefrontal cortex, hippocampus, and nucleus accumbens. Acute treatment with LY379268 restored NMDA-R and GABAA-R levels in the two-hit mouse model comparable to olanzapine. CONCLUSIONS We demonstrate that the mGluR2/3 agonist LY379268 restores excitatory and inhibitory deficits with similar efficiency as olanzapine in our two-hit schizophrenia mouse model. This study significantly contributes to our understanding of the mechanisms underlying the therapeutic effects of LY379268 and supports the use of agents aimed at mGluR2/3.
Collapse
Affiliation(s)
- Martin Engel
- Schizophrenia Research Institute, Sydney, Australia. .,Faculty of Science Medicine and Health, University of Wollongong, Wollongong, Australia. .,Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia. .,School of Biological Sciences, University of Wollongong, Wollongong, Australia.
| | - Peta Snikeris
- Schizophrenia Research Institute, Sydney, Australia.,Faculty of Science Medicine and Health, University of Wollongong, Wollongong, Australia.,Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Natalie Matosin
- Schizophrenia Research Institute, Sydney, Australia.,Faculty of Science Medicine and Health, University of Wollongong, Wollongong, Australia.,Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Kelly Anne Newell
- Schizophrenia Research Institute, Sydney, Australia.,Faculty of Science Medicine and Health, University of Wollongong, Wollongong, Australia.,Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Xu-Feng Huang
- Schizophrenia Research Institute, Sydney, Australia.,Faculty of Science Medicine and Health, University of Wollongong, Wollongong, Australia.,Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Elisabeth Frank
- Schizophrenia Research Institute, Sydney, Australia.,Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| |
Collapse
|
31
|
Witkin JM, Ornstein PL, Mitch CH, Li R, Smith SC, Heinz BA, Wang XS, Xiang C, Carter JH, Anderson WH, Li X, Broad LM, Pasqui F, Fitzjohn SM, Sanger HE, Smith JL, Catlow J, Swanson S, Monn JA. In vitro pharmacological and rat pharmacokinetic characterization of LY3020371, a potent and selective mGlu 2/3 receptor antagonist. Neuropharmacology 2015; 115:100-114. [PMID: 26748052 DOI: 10.1016/j.neuropharm.2015.12.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/09/2015] [Accepted: 12/22/2015] [Indexed: 12/27/2022]
Abstract
Metabotropic glutamate 2/3 (mGlu2/3) receptors are of considerable interest owing to their role in modulating glutamate transmission via presynaptic, postsynaptic and glial mechanisms. As part of our ongoing efforts to identify novel ligands for these receptors, we have discovered (1S,2R,3S,4S,5R,6R)-2-amino-3-[(3,4-difluorophenyl)sulfanylmethyl]-4-hydroxy-bicyclo[3.1.0]hexane-2,6-dicarboxylic acid; (LY3020371), a potent and selective orthosteric mGlu2/3 receptor antagonist. In this account, we characterize the effects of LY3020371 in membranes and cells expressing human recombinant mGlu receptor subtypes as well as in native rodent and human brain tissue preparations, providing important translational information for this molecule. In membranes from cells expressing recombinant human mGlu2 and mGlu3 receptor subtypes, LY3020371.HCl competitively displaced binding of the mGlu2/3 agonist ligand [3H]-459477 with high affinity (hmGlu2 Ki = 5.26 nM; hmGlu3 Ki = 2.50 nM). In cells expressing hmGlu2 receptors, LY3020371.HCl potently blocked mGlu2/3 agonist (DCG-IV)-inhibited, forskolin-stimulated cAMP formation (IC50 = 16.2 nM), an effect that was similarly observed in hmGlu3-expressing cells (IC50 = 6.21 nM). Evaluation of LY3020371 in cells expressing the other human mGlu receptor subtypes revealed high mGlu2/3 receptor selectivity. In rat native tissue assays, LY3020371 demonstrated effective displacement of [3H]-459477 from frontal cortical membranes (Ki = 33 nM), and functional antagonist activity in cortical synaptosomes measuring both the reversal of agonist-suppressed second messenger production (IC50 = 29 nM) and agonist-inhibited, K+-evoked glutamate release (IC50 = 86 nM). Antagonism was fully recapitulated in both primary cultured cortical neurons where LY3020371 blocked agonist-suppressed spontaneous Ca2+ oscillations (IC50 = 34 nM) and in an intact hippocampal slice preparation (IC50 = 46 nM). Functional antagonist activity was similarly demonstrated in synaptosomes prepared from epileptic human cortical or hippocampal tissues, suggesting a translation of the mGlu2/3 antagonist pharmacology from rat to human. Intravenous dosing of LY3020371 in rats led to cerebrospinal fluid drug levels that are expected to effectively block mGlu2/3 receptors in vivo. Taken together, these results establish LY3020371 as an important new pharmacological tool for studying mGlu2/3 receptors in vitro and in vivo. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Paul L Ornstein
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Charles H Mitch
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Renhua Li
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Stephon C Smith
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Beverly A Heinz
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Xu-Shan Wang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Chuanxi Xiang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Joan H Carter
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Wesley H Anderson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Xia Li
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | | | | | | | | | | | - John Catlow
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Steven Swanson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - James A Monn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA.
| |
Collapse
|
32
|
Monn JA, Prieto L, Taboada L, Hao J, Reinhard MR, Henry SS, Beadle CD, Walton L, Man T, Rudyk H, Clark B, Tupper D, Baker SR, Lamas C, Montero C, Marcos A, Blanco J, Bures M, Clawson DK, Atwell S, Lu F, Wang J, Russell M, Heinz BA, Wang X, Carter JH, Getman BG, Catlow JT, Swanson S, Johnson BG, Shaw DB, McKinzie DL. Synthesis and Pharmacological Characterization of C4-(Thiotriazolyl)-substituted-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylates. Identification of (1R,2S,4R,5R,6R)-2-Amino-4-(1H-1,2,4-triazol-3-ylsulfanyl)bicyclo[3.1.0]hexane-2,6-dicarboxylic Acid (LY2812223), a Highly Potent, Functionally Selective mGlu2 Receptor Agonist. J Med Chem 2015; 58:7526-48. [PMID: 26313429 DOI: 10.1021/acs.jmedchem.5b01124] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Identification of orthosteric mGlu(2/3) receptor agonists capable of discriminating between individual mGlu2 and mGlu3 subtypes has been highly challenging owing to the glutamate-site sequence homology between these proteins. Herein we detail the preparation and characterization of a series of molecules related to (1S,2S,5R,6S)-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylate 1 (LY354740) bearing C4-thiotriazole substituents. On the basis of second messenger responses in cells expressing other recombinant human mGlu2/3 subtypes, a number of high potency and efficacy mGlu2 receptor agonists exhibiting low potency mGlu3 partial agonist/antagonist activity were identified. From this, (1R,2S,4R,5R,6R)-2-amino-4-(1H-1,2,4-triazol-3-ylsulfanyl)bicyclo[3.1.0]hexane-2,6-dicarboxylic acid 14a (LY2812223) was further characterized. Cocrystallization of 14a with the amino terminal domains of hmGlu2 and hmGlu3 combined with site-directed mutation studies has clarified the underlying molecular basis of this unique pharmacology. Evaluation of 14a in a rat model responsive to mGlu2 receptor activation coupled with a measure of central drug disposition provides evidence that this molecule engages and activates central mGlu2 receptors in vivo.
Collapse
Affiliation(s)
- James A Monn
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Lourdes Prieto
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Lorena Taboada
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Junliang Hao
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Matthew R Reinhard
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Steven S Henry
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Christopher D Beadle
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Lesley Walton
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Teresa Man
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Helene Rudyk
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Barry Clark
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - David Tupper
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - S Richard Baker
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Carlos Lamas
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Carlos Montero
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Alicia Marcos
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Jaime Blanco
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Mark Bures
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - David K Clawson
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Shane Atwell
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Frances Lu
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Jing Wang
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Marijane Russell
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Beverly A Heinz
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Xushan Wang
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Joan H Carter
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Brian G Getman
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - John T Catlow
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Steven Swanson
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - Bryan G Johnson
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - David B Shaw
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| | - David L McKinzie
- Discovery Chemistry Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition and ⊥Neuroscience Research, Eli Lilly and Company , Lilly Corporate Center, Drop 0510, Indianapolis, Indiana 46285, United States
| |
Collapse
|
33
|
mGlu2 Receptor Agonism, but Not Positive Allosteric Modulation, Elicits Rapid Tolerance towards Their Primary Efficacy on Sleep Measures in Rats. PLoS One 2015; 10:e0144017. [PMID: 26658273 PMCID: PMC4684355 DOI: 10.1371/journal.pone.0144017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 11/12/2015] [Indexed: 12/17/2022] Open
Abstract
G-protein-coupled receptor (GPCR) agonists are known to induce both cellular adaptations resulting in tolerance to therapeutic effects and withdrawal symptoms upon treatment discontinuation. Glutamate neurotransmission is an integral part of sleep-wake mechanisms, which processes have translational relevance for central activity and target engagement. Here, we investigated the efficacy and tolerance potential of the metabotropic glutamate receptors (mGluR2/3) agonist LY354740 versus mGluR2 positive allosteric modulator (PAM) JNJ-42153605 on sleep-wake organisation in rats. In vitro, the selectivity and potency of JNJ-42153605 were characterized. In vivo, effects on sleep measures were investigated in rats after once daily oral repeated treatment for 7 days, withdrawal and consecutive re-administration of LY354740 (1–10 mg/kg) and JNJ-42153605 (3–30 mg/kg). JNJ-42153605 showed high affinity, potency and selectivity at mGluR2. Binding site analyses and knowledge-based docking confirmed the specificity of JNJ-42153605 at the mGluR2 allosteric binding site. Acute LY354740 and JNJ-42153605 dose-dependently decreased rapid eye movement (REM) sleep time and prolonged its onset latency. Sub chronic effects of LY354740 on REM sleep measures disappeared from day 3 onwards, whereas those of JNJ-42153605 were maintained after repeated exposure. LY354740 attenuated REM sleep homeostatic recovery, while this was preserved after JNJ-42153605 administration. JNJ-42153605 enhanced sleep continuity and efficiency, suggesting its potential as an add-on medication for impaired sleep quality during early stages of treatment. Abrupt cessation of JNJ-42153605 did not induce withdrawal phenomena and sleep disturbances, while the initial drug effect was fully reinstated after re-administration. Collectively, long-term treatment with JNJ-42153605 did not induce tolerance phenomena to its primary functional effects on sleep measures, nor adverse effects at withdrawal, while it promoted homeostatic recovery sleep. From the translational perspective, the present rodent findings suggest that mGluR2 positive allosteric modulation has therapeutic potential based on its superior long term efficacy over agonists in psychiatric disorders, particularly of those commonly occurring with REM sleep overdrive.
Collapse
|
34
|
In the grey zone between epilepsy and schizophrenia: alterations in group II metabotropic glutamate receptors. Acta Neurol Belg 2015; 115:221-32. [PMID: 25539775 DOI: 10.1007/s13760-014-0407-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/05/2014] [Indexed: 01/09/2023]
Abstract
Glutamate is the major excitatory neurotransmitter in the brain. The glutamate system plays an important role in the formation of synapses during brain development and synaptic plasticity. Dysfunctions in glutamate regulation may lead to hyperexcitatory neuronal networks and neurotoxicity. Glutamate excess is possibly of great importance in the pathophysiology of several neurological and psychiatric disorders such as epilepsy and schizophrenia. Interestingly, cross talk between these disorders has been well documented: psychiatric comorbidities are frequent in epilepsy and temporal lobe epilepsy is one of the highest risk factors for developing psychosis. Therefore, dysfunctions in glutamatergic neurotransmission might constitute a common pathological mechanism. A major negative feedback system is regulated by the presynaptic group II metabotropic glutamate (mGlu) receptors including mGlu2/3 receptors. These receptors are predominantly localised extrasynaptically in basal ganglia and limbic structures. Hence, mGlu2/3 receptors are an interesting target for the treatment of disorders like epilepsy and schizophrenia. A dysfunction in the glutamate system may be associated with alterations in mGlu2/3 receptor expression. In this review, we describe the localization of mGlu2/3 receptors in the healthy brain of mice, rats and humans. Secondly, changes in mGlu2/3 receptor density of the brain regions affected in epilepsy and schizophrenia are summarised. Increased mGlu2/3 receptor density might represent a compensatory mechanism of the brain to regulate elevated glutamate levels, while reduced mGlu2/3 receptor density in some brain regions may further contribute to the aberrant hyperexcitability. Further research considering the mGlu2/3 receptor can contribute significantly to the understanding of the etiological and therapeutic role of group II mGlu receptor in epilepsy, epilepsy with psychosis and schizophrenia.
Collapse
|
35
|
Pritchett D, Jagannath A, Brown LA, Tam SKE, Hasan S, Gatti S, Harrison PJ, Bannerman DM, Foster RG, Peirson SN. Deletion of Metabotropic Glutamate Receptors 2 and 3 (mGlu2 & mGlu3) in Mice Disrupts Sleep and Wheel-Running Activity, and Increases the Sensitivity of the Circadian System to Light. PLoS One 2015; 10:e0125523. [PMID: 25950516 PMCID: PMC4423919 DOI: 10.1371/journal.pone.0125523] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/14/2015] [Indexed: 12/22/2022] Open
Abstract
Sleep and/or circadian rhythm disruption (SCRD) is seen in up to 80% of schizophrenia patients. The co-morbidity of schizophrenia and SCRD may in part stem from dysfunction in common brain mechanisms, which include the glutamate system, and in particular, the group II metabotropic glutamate receptors mGlu2 and mGlu3 (encoded by the genes Grm2 and Grm3). These receptors are relevant to the pathophysiology and potential treatment of schizophrenia, and have also been implicated in sleep and circadian function. In the present study, we characterised the sleep and circadian rhythms of Grm2/3 double knockout (Grm2/3-/-) mice, to provide further evidence for the involvement of group II metabotropic glutamate receptors in the regulation of sleep and circadian rhythms. We report several novel findings. Firstly, Grm2/3-/- mice demonstrated a decrease in immobility-determined sleep time and an increase in immobility-determined sleep fragmentation. Secondly, Grm2/3-/- mice showed heightened sensitivity to the circadian effects of light, manifested as increased period lengthening in constant light, and greater phase delays in response to nocturnal light pulses. Greater light-induced phase delays were also exhibited by wildtype C57Bl/6J mice following administration of the mGlu2/3 negative allosteric modulator RO4432717. These results confirm the involvement of group II metabotropic glutamate receptors in photic entrainment and sleep regulation pathways. Finally, the diurnal wheel-running rhythms of Grm2/3-/- mice were perturbed under a standard light/dark cycle, but their diurnal rest-activity rhythms were unaltered in cages lacking running wheels, as determined with passive infrared motion detectors. Hence, when assessing the diurnal rest-activity rhythms of mice, the choice of assay can have a major bearing on the results obtained.
Collapse
Affiliation(s)
- David Pritchett
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Aarti Jagannath
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- F.Hoffman-La Roche, Neuroscience, Ophthalmology & Rare Diseases (NORD), Pharma Research & Early Development (pRED) Innovation Centre, Basel, Switzerland
| | - Laurence A. Brown
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Shu K. E. Tam
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Sibah Hasan
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Silvia Gatti
- F.Hoffman-La Roche, Neuroscience, Ophthalmology & Rare Diseases (NORD), Pharma Research & Early Development (pRED) Innovation Centre, Basel, Switzerland
| | - Paul J. Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, United Kingdom
| | - David M. Bannerman
- Department of Experimental Psychology, University of Oxford, South Parks Road, Oxford, OX1 3UD, United Kingdom
| | - Russell G. Foster
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- * E-mail: (RGF); (SNP)
| | - Stuart N. Peirson
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- * E-mail: (RGF); (SNP)
| |
Collapse
|
36
|
Salih H, Anghelescu I, Kezic I, Sinha V, Hoeben E, Van Nueten L, De Smedt H, De Boer P. Pharmacokinetic and pharmacodynamic characterisation of JNJ-40411813, a positive allosteric modulator of mGluR2, in two randomised, double-blind phase-I studies. J Psychopharmacol 2015; 29:414-25. [PMID: 25735992 DOI: 10.1177/0269881115573403] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Metabotropic glutamate receptor-2 positive allosteric modulator, JNJ-40411813 (ADX71149), was characterised for clinical effects in healthy volunteers in two phase-1 studies. In study 1, healthy men received 50-, 100-, 150- or 225 mg and women received 100 mg JNJ-40411813 (n=6, each cohort) or placebo (n=2, each cohort) twice daily for seven days; smoking men (n=30) received placebo twice daily on days 1-7, 100 mg JNJ-40411813 (n=20) or placebo (n=10) on days 8-14. In study 2, healthy men received intravenous 0.005 mg/kg S(+) ketamine over 60 min at 3 (n=24; cohort 1), 12 h (n=8; cohort 3), and 24 h (n=8; cohort 2) after a single oral dose of 500 mg JNJ-40411813 or placebo. The pharmacokinetics and effects of JNJ-40411813 on cognition and subjective awareness were evaluated. Plasma JNJ-40411813 exposure was dose-dependent, t max ranged from 3-4 h and t 1/2 19.4-34.2 h across the dose levels. JNJ-40411813 significantly (p=0.02) reduced continuity of attention score (150 mg dose) and ameliorated smoking withdrawal-induced changes in power of attention and quality of episodic memory versus placebo. A modest reduction in alertness was observed at 150-225 mg doses, JNJ-40411813 (500 mg) reduced S(+) ketamine-induced negative symptoms by approximately 43% and 30% in cohorts 1 and 3, respectively. JNJ-40411813 was generally well-tolerated.
Collapse
Affiliation(s)
- Hiba Salih
- Janssen Research & Development, Beerse, Belgium
| | | | - Iva Kezic
- Janssen Research & Development, Beerse, Belgium
| | | | - Eef Hoeben
- Janssen Research & Development, Beerse, Belgium
| | | | | | | |
Collapse
|
37
|
Zhao C, Gammie SC. Metabotropic glutamate receptor 3 is downregulated and its expression is shifted from neurons to astrocytes in the mouse lateral septum during the postpartum period. J Histochem Cytochem 2015; 63:417-26. [PMID: 25739438 DOI: 10.1369/0022155415578283] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 02/27/2015] [Indexed: 12/11/2022] Open
Abstract
The inhibitory metabotropic glutamate receptor 3 (mGluR3) plays diverse and complex roles in brain function, including synaptic plasticity and neurotransmission. We recently found that mGluR3 is downregulated in the lateral septum (LS) of postpartum females using microarray and qPCR analysis. In this study, we used double fluorescence immunohistochemical approaches to characterize mGluR3 changes in LS of the postpartum brain. The number of mGluR3-immunoractive cells was significantly reduced in the dorsal (LSD) and intermediate (LSI) but not ventral (LSV) parts of the LS in postpartum versus virgin females. mGluR3 immunoreactivity in the LS was found predominantly in neurons (~70%), with a smaller portion (~20%-30%) in astrocytes. Colocalization analysis revealed a reduced mGluR3 expression in neurons but an increased astrocytic localization in postpartum LSI. This change in the pattern of expression suggests that mGluR3 expression is shifted from neurons to astrocytes in postpartum LS, and the decrease in mGluR3 is neuron-specific. Because mGluR3 is inhibitory and negatively regulates glutamate and GABA release, decreases in neuronal expression would increase glutamate and GABA signaling. Given our recent finding that ~90% of LS neurons are GABAergic, the present data suggest that decreases in mGluR3 are a mechanism for elevated GABA in LS in the postpartum state.
Collapse
Affiliation(s)
- Changjiu Zhao
- Department of Zoology, University of Wisconsin-Madison, Madison, Wisconsin (CZ, SCG)
| | - Stephen C Gammie
- Department of Zoology, University of Wisconsin-Madison, Madison, Wisconsin (CZ, SCG),Neuroscience Training Program, University of Wisconsin-Madison, Madison, Wisconsin (SCG)
| |
Collapse
|
38
|
Farinha A, Lavreysen H, Peeters L, Russo B, Masure S, Trabanco AA, Cid J, Tresadern G. Molecular determinants of positive allosteric modulation of the human metabotropic glutamate receptor 2. Br J Pharmacol 2015; 172:2383-96. [PMID: 25571949 DOI: 10.1111/bph.13065] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 12/20/2014] [Accepted: 12/23/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE The activation of the metabotropic glutamate receptor 2 (mGlu2 ) reduces glutamatergic transmission in brain regions where excess excitatory signalling is implicated in disorders such as anxiety and schizophrenia. Positive allosteric modulators (PAMs) can provide a fine-tuned potentiation of these receptors' function and are being investigated as a novel therapeutic approach. An extensive set of mutant human mGlu2 receptors were used to investigate the molecular determinants that are important for positive allosteric modulation at this receptor. EXPERIMENTAL APPROACH Site-directed mutagenesis, binding and functional assays were employed to identify amino acids important for the activity of nine PAMs. The data from the radioligand binding and mutagenesis studies were used with computational docking to predict a binding mode at an mGlu2 receptor model based on the recent structure of the mGlu1 receptor. KEY RESULTS New amino acids in TM3 (R635, L639, F643), TM5 (L732) and TM6 (W773, F776) were identified for the first time as playing an important role in the activity of mGlu2 PAMs. CONCLUSIONS AND IMPLICATIONS This extensive study furthers our understanding of positive allosteric modulation of the mGlu2 receptor and can contribute to improved future design of mGlu2 PAMs.
Collapse
Affiliation(s)
- A Farinha
- Neuroscience Discovery, Janssen Research and Development, Division of Janssen Pharmaceutica, Beerse, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Urstadt KR, Stanley BG. Direct hypothalamic and indirect trans-pallidal, trans-thalamic, or trans-septal control of accumbens signaling and their roles in food intake. Front Syst Neurosci 2015; 9:8. [PMID: 25741246 PMCID: PMC4327307 DOI: 10.3389/fnsys.2015.00008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 01/15/2015] [Indexed: 01/01/2023] Open
Abstract
Due in part to the increasing incidence of obesity in developed nations, recent research aims to elucidate neural circuits that motivate humans to overeat. Earlier research has described how the nucleus accumbens shell (AcbSh) motivates organisms to feed by activating neuronal populations in the lateral hypothalamus (LH). However, more recent research suggests that the LH may in turn communicate with the AcbSh, both directly and indirectly, to re-tune the motivation to consume foods with homeostatic and food-related sensory signals. Here, we discuss the functional and anatomical evidence for an LH to AcbSh connection and its role in eating behaviors. The LH appears to modulate Acb activity directly, using neurotransmitters such as hypocretin/orexin or melanin concentrating hormone (MCH). The LH also indirectly regulates AcbSh activity through certain subcortical "relay" regions, such as the lateral septum (LS), ventral pallidum (VP), and paraventricular thalamus, using a variety of neurotransmitters. This review aims to summarize studies on these topics and outline a model by which LH circuits processing energy balance can modulate AcbSh neural activity to regulate feeding behavior.
Collapse
Affiliation(s)
- Kevin R Urstadt
- Department of Psychology, University of Michigan Ann Arbor, MI, USA
| | - B Glenn Stanley
- Departments of Psychology and Cell Biology and Neuroscience, University of California - Riverside Riverside, CA, USA
| |
Collapse
|
40
|
Monn JA, Prieto L, Taboada L, Pedregal C, Hao J, Reinhard MR, Henry SS, Goldsmith PJ, Beadle CD, Walton L, Man T, Rudyk H, Clark B, Tupper D, Baker SR, Lamas C, Montero C, Marcos A, Blanco J, Bures M, Clawson DK, Atwell S, Lu F, Wang J, Russell M, Heinz BA, Wang X, Carter JH, Xiang C, Catlow JT, Swanson S, Sanger H, Broad LM, Johnson MP, Knopp KL, Simmons RMA, Johnson BG, Shaw DB, McKinzie DL. Synthesis and Pharmacological Characterization of C4-Disubstituted Analogs of 1S,2S,5R,6S-2-Aminobicyclo[3.1.0]hexane-2,6-dicarboxylate: Identification of a Potent, Selective Metabotropic Glutamate Receptor Agonist and Determination of Agonist-Bound Human mGlu2 and mGlu3 Amino Terminal Domain Structures. J Med Chem 2015; 58:1776-94. [DOI: 10.1021/jm501612y] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- James A. Monn
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Lourdes Prieto
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Lorena Taboada
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Concepcion Pedregal
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Junliang Hao
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Matt R. Reinhard
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Steven S. Henry
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Paul J. Goldsmith
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Christopher D. Beadle
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Lesley Walton
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Teresa Man
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Helene Rudyk
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Barry Clark
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - David Tupper
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - S. Richard Baker
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Carlos Lamas
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Carlos Montero
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Alicia Marcos
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Jaime Blanco
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Mark Bures
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - David K. Clawson
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Shane Atwell
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Frances Lu
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Jing Wang
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Marijane Russell
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Beverly A. Heinz
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Xushan Wang
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Joan H. Carter
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Chuanxi Xiang
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - John T. Catlow
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Steven Swanson
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Helen Sanger
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Lisa M. Broad
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Michael P. Johnson
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Kelly L. Knopp
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Rosa M. A. Simmons
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Bryan G. Johnson
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - David B. Shaw
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - David L. McKinzie
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| |
Collapse
|
41
|
Pratt JA, Morris BJ. The thalamic reticular nucleus: a functional hub for thalamocortical network dysfunction in schizophrenia and a target for drug discovery. J Psychopharmacol 2015; 29:127-37. [PMID: 25586397 DOI: 10.1177/0269881114565805] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The thalamus (comprising many distinct nuclei) plays a key role in facilitating sensory discrimination and cognitive processes through connections with the cortex. Impaired thalamocortical processing has long been considered to be involved in schizophrenia. In this review we focus on the thalamic reticular nucleus (TRN) providing evidence for it being an important communication hub between the thalamus and cortex and how it may play a key role in the pathophysiology of schizophrenia. We first highlight the functional neuroanatomy, neurotransmitter localisation and physiology of the TRN. We then present evidence of the physiological roles of the TRN in relation to oscillatory activity, cognition and behaviour. Next we discuss the role of the TRN in rodent models of risk factors for schizophrenia (genetic and pharmacological) and provide evidence for TRN deficits in schizophrenia. Finally we discuss new drug targets for schizophrenia in relation to restoring TRN circuitry dysfunction.
Collapse
Affiliation(s)
- Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK Psychiatric Research Institute of Neuroscience in Glasgow (PsyRING), University of Glasgow and University of Strathclyde, Glasgow, UK
| | - Brian J Morris
- Psychiatric Research Institute of Neuroscience in Glasgow (PsyRING), University of Glasgow and University of Strathclyde, Glasgow, UK Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
42
|
Copeland CS, Neale SA, Salt TE. Neuronal activity patterns in the mediodorsal thalamus and related cognitive circuits are modulated by metabotropic glutamate receptors. Neuropharmacology 2015; 92:16-24. [PMID: 25576798 PMCID: PMC4362770 DOI: 10.1016/j.neuropharm.2014.12.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/02/2014] [Accepted: 12/26/2014] [Indexed: 01/03/2023]
Abstract
The mediodorsal thalamus (MD) likely plays an important role in cognition as it receives abundant afferent connections from the amygdala and prefrontal cortex (PFC). Indeed, disturbed activity within the MD is thought to precipitate cognitive deficits associated with schizophrenia. As compounds acting at the Group II metabotropic glutamate (mGlu) receptors (subtypes mGlu2/mGlu3) have efficacy in animal models of schizophrenia, we investigated whether a Group II agonist and an mGlu2 positive allosteric modulator (PAM) could modulate MD activity. Extracellular single-unit recordings were made in vivo from MD neurones in anaesthetised rats. Responses were elicited by electrical stimulation of the PFC and/or amygdala, with Group II compounds locally applied as required. The Group II agonist reduced inhibition evoked in the MD: an effect manifested as an increase in short-latency responses, and a decrease in long-latency burst-firing. This disinhibitory action of the Group II receptors in the MD represents a mechanism of potential therapeutic importance as increased inhibition in the MD has been associated with cognitive deficit-onset. Furthermore, as co-application of the mGlu2 PAM did not potentiate the Group II agonist effects in the MD, we suggest that the Group II disinhibitory effect is majority-mediated via mGlu3. This heterogeneity in Group II receptor thalamic physiology bears consequence, as compounds active exclusively at the mGlu2 subtype are unlikely to perturb maladapted MD firing patterns associated with cognitive deficits, with activity at mGlu3 receptors possibly more appropriate. Indeed, polymorphisms in the mGlu3, but not the mGlu2, gene have been detected in patients with schizophrenia. There is heterogeneity in Group II receptor physiology across thalamic nuclei. This differential distribution may facilitate multimodal thalamic nuclei functions. Group II receptor activation reduced burst firing via reducing thalamic inhibition. Increased thalamic inhibition precipitates impairments in cognitive function. Activating the Group II receptors may therefore enhance cognitive function.
Collapse
Affiliation(s)
- C S Copeland
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - S A Neale
- Neurexpert Ltd, Kemp House, City Road, London, EC1V 2NX, UK.
| | - T E Salt
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
43
|
Celanire S, Sebhat I, Wichmann J, Mayer S, Schann S, Gatti S. Novel metabotropic glutamate receptor 2/3 antagonists and their therapeutic applications: a patent review (2005 - present). Expert Opin Ther Pat 2014; 25:69-90. [PMID: 25435285 DOI: 10.1517/13543776.2014.983899] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION This review focuses on the medicinal chemistry efforts directed toward the identification of competitive and noncompetitive antagonists of glutamate at group II metabotropic glutamate receptors (mGluRII: mGlu2/3 and mGlu2). This class of compounds holds promise for the treatment of CNS disorders such as major depression, cognitive deficits and sleep-wake disorders, and several pharmaceutical companies are advancing mGluRII antagonists from discovery research into clinical development. AREA COVERED This review article covers for the first time the patent applications that were published on mGlu2/3 orthosteric and allosteric antagonists between January 2005 and September 2014, with support from the primary literature, posters and oral communications from international congresses. Patent applications published prior to 2005 for which compositions of matter were largely described in peer review articles are briefly discussed with main findings. EXPERT OPINION Recent advances in the prodrug approach of novel mGlu2/3 orthosteric antagonists combined with the design of novel mGlu2/3 and mGlu2 negative allosteric modulators provide new therapeutic opportunities for neurologic and psychiatric disorders.
Collapse
Affiliation(s)
- Sylvain Celanire
- CEO, Pragma Therapeutics , 9 rue Ada Byron, Domaine de Chosal, Archamp Technopole, 74166 Saint-Julien-en-Genevois Cedex , France +33 6 79 85 37 06 ;
| | | | | | | | | | | |
Collapse
|
44
|
Risk genes for schizophrenia: Translational opportunities for drug discovery. Pharmacol Ther 2014; 143:34-50. [DOI: 10.1016/j.pharmthera.2014.02.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 01/31/2014] [Indexed: 12/11/2022]
|
45
|
Ahnaou A, Ver Donck L, Drinkenburg WHIM. Blockade of the metabotropic glutamate (mGluR2) modulates arousal through vigilance states transitions: evidence from sleep-wake EEG in rodents. Behav Brain Res 2014; 270:56-67. [PMID: 24821401 DOI: 10.1016/j.bbr.2014.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/15/2014] [Accepted: 05/02/2014] [Indexed: 02/03/2023]
Abstract
Accumulating data continue to support the therapeutic potential of glutamate metabotropic (mGluR2) receptors for treatment of psychiatric disorders such as depression, anxiety and schizophrenia. Glutamate neurotransmission is an integral component of sleep-wake mechanisms, which have translational relevance to assess on-target activity of drugs. Here, we investigated the influence of mGluR2 inactivation upon sleep-wake electroencephalogram (EEG) in rodents. Rats were administered with vehicle, the specific mGluR2 antagonist LY341495 (2.5, 5, 10mg/kg) or negative allosteric modulator (NAM) Ro-4491533 (2.5, 10 and 40 mg/kg) at lights onset. mGluR2 (-/-) mice were used to confirm the selectivity of functional response. Both LY341495 and Ro-4491533 induced an immediate and endured desynchronized cortical activity during 3-6h associated with enhanced theta and gamma oscillations and depressed slow oscillations during sleep. The arousal-promoting effect is consistent with the marked lengthening of sleep onset latency, an increased number of state transitions from light sleep to waking and the gradual increase in homeostatic compensatory sleep. The arousal response to mGluR2 blockade was not accompanied by sharp rebound hypersomnolence as found with the classical psycho-stimulant amphetamine. mGluR2 (-/-) mice and their WT littermates exhibited similar sleep-wake phenotype, while Ro-4491533 (40 mg/kg) enhanced waking associated with increased locomotor activity and body temperature in WT but not in mGluR2 (-/-) mice, which confirm the role of mGluR2 inactivation in the arousal response. Our results lend support for a role of mGluR2 blockade in promoting cortical arousal associated with theta/gamma oscillations as well as high thresholds transitions from sleep to waking.
Collapse
Affiliation(s)
- A Ahnaou
- Department of Neurosciences, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium.
| | - L Ver Donck
- Department of Neurosciences, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - W H I M Drinkenburg
- Department of Neurosciences, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
46
|
Kolaj M, Zhang L, Hermes MLHJ, Renaud LP. Intrinsic properties and neuropharmacology of midline paraventricular thalamic nucleus neurons. Front Behav Neurosci 2014; 8:132. [PMID: 24860449 PMCID: PMC4029024 DOI: 10.3389/fnbeh.2014.00132] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 04/01/2014] [Indexed: 01/01/2023] Open
Abstract
Neurons in the midline and intralaminar thalamic nuclei are components of an interconnected brainstem, limbic and prefrontal cortex neural network that is engaged during arousal, vigilance, motivated and addictive behaviors, and stress. To better understand the cellular mechanisms underlying these functions, here we review some of the recently characterized electrophysiological and neuropharmacological properties of neurons in the paraventricular thalamic nucleus (PVT), derived from whole cell patch clamp recordings in acute rat brain slice preparations. PVT neurons display firing patterns and ionic conductances (IT and IH) that exhibit significant diurnal change. Their resting membrane potential (RMP) is maintained by various ionic conductances that include inward rectifier (Kir), hyperpolarization-activated nonselective cation (HCN) and TWIK-related acid sensitive (TASK) K+ channels. Firing patterns are regulated by high voltage-activated (HVA) and low voltage-activated (LVA) Ca2+ conductances. Moreover, transient receptor potential (TRP)-like nonselective cation channels together with Ca2+- and Na+-activated K+ conductances (KCa; KNa) contribute to unique slow afterhyperpolarizing potentials (sAHPs) that are generally not detectable in lateral thalamic or reticular thalamic nucleus neurons. The excitability of PVT neurons is also modulated by activation of neurotransmitter receptors associated with afferent pathways to PVT and other thalamic midline nuclei. We report on receptor-mediated actions of GABA, glutamate, monoamines and several neuropeptides: arginine vasopressin, gastrin-releasing peptide, thyrotropin releasing hormone and the orexins (hypocretins). This review represents an initial survey of intrinsic and transmitter-sensitive ionic conductances that are deemed to be unique to this population of midline thalamic neurons, information that is fundamental to an appreciation of the role these thalamic neurons may play in normal central nervous system (CNS) physiology and in CNS disorders that involve the dorsomedial thalamus.
Collapse
Affiliation(s)
- Miloslav Kolaj
- Neuroscience Program and Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada
| | - Li Zhang
- Neuroscience Program and Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada
| | - Michael L H J Hermes
- Neuroscience Program and Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada
| | - Leo P Renaud
- Neuroscience Program and Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada
| |
Collapse
|
47
|
Lane TA, Boerner T, Bannerman DM, Kew JNC, Tunbridge EM, Sharp T, Harrison PJ. Decreased striatal dopamine in group II metabotropic glutamate receptor (mGlu2/mGlu3) double knockout mice. BMC Neurosci 2013; 14:102. [PMID: 24053122 PMCID: PMC3857325 DOI: 10.1186/1471-2202-14-102] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 09/17/2013] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Group II metabotropic glutamate receptors (mGlu2 and mGlu3, encoded by Grm2 and Grm3) have been the focus of attention as treatment targets for a number of psychiatric conditions. Double knockout mice lacking mGlu2 and mGlu3 (mGlu2/3-/-) show a subtle behavioural phenotype, being hypoactive under basal conditions and in response to amphetamine, and with a spatial memory deficit that depends on the arousal properties of the task. The neurochemical correlates of this profile are unknown. Here, we measured tissue levels of dopamine, 5-HT, noradrenaline and their metabolites in the striatum and frontal cortex of mGlu2/3-/- double knockout mice, using high performance liquid chromatography. We also measured the same parameters in mGlu2-/- and mGlu3-/- single knockout mice. RESULTS mGlu2/3-/-mice had reduced dopamine levels in the striatum but not in frontal cortex, compared to wild-types. In a separate cohort we replicated this deficit and, using tissue punches, found it was more prominent in the nucleus accumbens than in dorsolateral striatum. Noradrenaline, 5-HT and their metabolites were not altered in the striatum of mGlu2/3-/- mice, although the noradrenaline metabolite MHPG was increased in the cortex. In mGlu2-/- and mGlu3-/- single knockout mice we found no difference in any monoamine or metabolite, in either brain region, compared to their wild-type littermates. CONCLUSIONS Group II metabotropic glutamate receptors impact upon striatal dopamine. The effect may contribute to the behavioural phenotype of mGlu2/3-/- mice. The lack of dopaminergic alterations in mGlu2-/- and mGlu3-/- single knockout mice reveals a degree of redundancy between the two receptors. The findings support the possibility that interactions between mGlu2/3 and dopamine may be relevant to the pathophysiology and therapy of schizophrenia and other disorders.
Collapse
Affiliation(s)
- Tracy A Lane
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK.
| | | | | | | | | | | | | |
Collapse
|
48
|
Wierońska JM, Sławińska A, Stachowicz K, Łasoń-Tyburkiewicz M, Gruca P, Papp M, Pilc A. The reversal of cognitive, but not negative or positive symptoms of schizophrenia, by the mGlu₂/₃ receptor agonist, LY379268, is 5-HT₁A dependent. Behav Brain Res 2013; 256:298-304. [PMID: 23948211 DOI: 10.1016/j.bbr.2013.08.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/26/2013] [Accepted: 08/02/2013] [Indexed: 11/24/2022]
Abstract
mGlu(2/3) receptor agonists were shown to possess an antipsychotic-like potential in animal studies. Recent clinical investigations revealed that their antipsychotic potential might also manifest in humans. LY379268, the group II mGlu receptor orthosteric agonist, was previously shown to exhibit antipsychotic-like action in animal models of schizophrenia. However, the mechanism of its action is not fully recognized. Here, we decided to investigate the involvement of 5-HT1A receptors in the LY379268-induced antipsychotic effects. We used models of positive, negative and cognitive symptoms of schizophrenia, such as MK-801- and amphetamine-induced hyperactivity tests, DOI-induced head twitches, social interaction and novel object recognition. LY379268 was active in a wide range of doses (0.5-5 mg/kg), depending on the paradigm. The effects of the drug were not antagonized by 5-HT(1A) antagonist, WAY100635 (0.1 mg/kg) in the models of positive and negative symptoms. Conversely, in the novel object recognition test, which exerts cognitive disturbances, the action of LY379268 was antagonized by WAY100635. Concomitantly, the action of a sub-effective dose of the drug was enhanced by the administration of a sub-effective dose of 5-HT(1A) agonist, (R)-(+)-8-Hydroxy-DPAT. Altogether, we propose that the antipsychotic-like action of group II mGlu receptors' agonist is 5-HT(1A) independent in context of positive and negative symptoms, while the action toward cognitive disturbances seems to be 5-HT(1A) dependent.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland.
| | | | | | | | | | | | | |
Collapse
|
49
|
Lavreysen H, Langlois X, Ahnaou A, Drinkenburg W, te Riele P, Biesmans I, Van der Linden I, Peeters L, Megens A, Wintmolders C, Cid JM, Trabanco AA, Andrés JI, Dautzenberg FM, Lütjens R, Macdonald G, Atack JR. Pharmacological characterization of JNJ-40068782, a new potent, selective, and systemically active positive allosteric modulator of the mGlu2 receptor and its radioligand [3H]JNJ-40068782. J Pharmacol Exp Ther 2013; 346:514-27. [PMID: 23766542 DOI: 10.1124/jpet.113.204990] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Modulation of the metabotropic glutamate type 2 (mGlu2) receptor is considered a promising target for the treatment of central nervous system diseases such as schizophrenia. Here, we describe the pharmacological properties of the novel mGlu2 receptor positive allosteric modulator (PAM) 3-cyano-1-cyclopropylmethyl-4-(4-phenyl-piperidin-1-yl)-pyridine-2(1H)-one (JNJ-40068782) and its radioligand [(3)H]JNJ-40068782. In guanosine 5'-O-(3-[(35)S]thio)triphosphate binding, JNJ-40068782 produced a leftward and upward shift in the glutamate concentration-effect curve at human recombinant mGlu2 receptors. The EC50 of JNJ-40068782 for potentiation of an EC20-equivalent concentration of glutamate was 143 nM. Although JNJ-40068782 did not affect binding of the orthosteric antagonist [(3)H]2S-2-amino-2-(1S,2S-2-carboxycyclopropyl-1-yl)-3-(xanth-9-yl)propanoic acid (LY-341495), it did potentiate the binding of the agonist [(3)H](2S,2'R,3'R)-2-(2',3'-dicarboxylcyclopropyl)glycine (DCG-IV), demonstrating that it can allosterically affect binding at the agonist recognition site. The binding of [(3)H]JNJ-40068782 to human recombinant mGlu2 receptors in Chinese hamster ovary cells and rat brain receptors was saturable with a KD of ∼10 nM. In rat brain, the anatomic distribution of [(3)H]JNJ-40068782 was consistent with mGlu2 expression previously described and was most abundant in cortex and hippocampus. The ability of structurally unrelated PAMs to displace [(3)H]JNJ-40068782 suggests that PAMs may bind to common determinants within the same site. It is noteworthy that agonists also increased the binding affinity of [(3)H]JNJ-40068782. JNJ-40068782 influenced rat sleep-wake organization by decreasing rapid eye movement sleep with a lowest active dose of 3 mg/kg PO. In mice, JNJ-40068782 reversed phencyclidine-induced hyperlocomotion with an ED50 of 5.7 mg/kg s.c. Collectively, the present data demonstrate that JNJ-40068782 has utility in investigating the potential of mGlu2 modulation for the treatment of diseases characterized by disturbed glutamatergic signaling and highlight the value of [(3)H]JNJ-40068782 in exploring allosteric binding.
Collapse
|
50
|
Ye ZY, Li DP, Pan HL. Regulation of Hypothalamic Presympathetic Neurons and Sympathetic Outflow by Group II Metabotropic Glutamate Receptors in Spontaneously Hypertensive Rats. Hypertension 2013; 62:255-62. [PMID: 23716583 DOI: 10.1161/hypertensionaha.113.01466] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Increased glutamatergic input in the hypothalamic paraventricular nucleus (PVN) plays an important role in the development of hypertension. Group II metabotropic glutamate receptors are expressed in the PVN, but their involvement in regulating synaptic transmission and sympathetic outflow in hypertension is unclear. Here, we show that the group II metabotropic glutamate receptors agonist (2S,2'R,3'R)-2-(2',3'-dicarboxycyclopropyl)glycine (DCG-IV) produced a significantly greater reduction in the frequency of spontaneous and miniature excitatory postsynaptic currents and in the amplitude of electrically evoked excitatory postsynaptic currents in retrogradely labeled spinally projecting PVN neurons in spontaneously hypertensive rats (SHRs) than in normotensive control rats. DCG-IV similarly decreased the frequency of GABAergic inhibitory postsynaptic currents of labeled PVN neurons in the 2 groups of rats. Strikingly, DCG-IV suppressed the firing of labeled PVN neurons only in SHRs. DCG-IV failed to inhibit the firing of PVN neurons of SHRs in the presence of ionotropic glutamate receptor antagonists. Lowering blood pressure with celiac ganglionectomy in SHRs normalized the DCG-IV effect on excitatory postsynaptic currents to the same level seen in control rats. Furthermore, microinjection of DCG-IV into the PVN significantly reduced blood pressure and sympathetic nerve activity in SHRs. Our findings provide new information that presynaptic group II metabotropic glutamate receptor activity at the glutamatergic terminals increases in the PVN in SHRs. Activation of group II metabotropic glutamate receptors in the PVN inhibits sympathetic vasomotor tone through attenuation of increased glutamatergic input and neuronal hyperactivity in SHRs.
Collapse
Affiliation(s)
- Zeng-You Ye
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030-4009, USA
| | | | | |
Collapse
|