1
|
Domin H, Burnat G. mGlu4R, mGlu7R, and mGlu8R allosteric modulation for treating acute and chronic neurodegenerative disorders. Pharmacol Rep 2024; 76:1219-1241. [PMID: 39348087 PMCID: PMC11582148 DOI: 10.1007/s43440-024-00657-7] [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: 07/19/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024]
Abstract
Neuroprotection, defined as safeguarding neurons from damage and death by inhibiting diverse pathological mechanisms, continues to be a promising approach for managing a range of central nervous system (CNS) disorders, including acute conditions such as ischemic stroke and traumatic brain injury (TBI) and chronic neurodegenerative diseases like Parkinson's disease (PD), Alzheimer's disease (AD), and multiple sclerosis (MS). These pathophysiological conditions involve excessive glutamatergic (Glu) transmission activity, which can lead to excitotoxicity. Inhibiting this excessive Glu transmission has been proposed as a potential therapeutic strategy for treating the CNS disorders mentioned. In particular, ligands of G protein-coupled receptors (GPCRs), including metabotropic glutamatergic receptors (mGluRs), have been recognized as promising options for inhibiting excessive Glu transmission. This review discusses the complex interactions of mGlu receptors with their subtypes, including the formation of homo- and heterodimers, which may vary in function and pharmacology depending on their protomer composition. Understanding these intricate details of mGlu receptor structure and function enhances researchers' ability to develop targeted pharmacological interventions, potentially offering new therapeutic avenues for neurological and psychiatric disorders. This review also summarizes the current knowledge of the neuroprotective potential of ligands targeting group III mGluRs in preclinical cellular (in vitro) and animal (in vivo) models of ischemic stroke, TBI, PD, AD, and MS. In recent years, experiments have shown that compounds, especially those activating mGlu4 or mGlu7 receptors, exhibit protective effects in experimental ischemia models. The discovery of allosteric ligands for specific mGluR subtypes has led to reports suggesting that group III mGluRs may be promising targets for neuroprotective therapy in PD (mGlu4R), TBI (mGlu7R), and MS (mGlu8R).
Collapse
Affiliation(s)
- Helena Domin
- Maj Institute of Pharmacology, Department of Neurobiology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland.
| | - Grzegorz Burnat
- Maj Institute of Pharmacology, Department of Neurobiology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland
| |
Collapse
|
2
|
Ramsakha N, Ojha P, Pal S, Routh S, Citri A, Bhattacharyya S. A vital role for PICK1 in the differential regulation of metabotropic glutamate receptor internalization and synaptic AMPA receptor endocytosis. J Biol Chem 2023:104837. [PMID: 37209824 DOI: 10.1016/j.jbc.2023.104837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/19/2023] [Accepted: 05/12/2023] [Indexed: 05/22/2023] Open
Abstract
Group I metabotropic glutamate receptors (mGluRs) play important roles in many neuronal processes and are believed to be involved in synaptic plasticity underlying the encoding of experience, including classic paradigms of learning and memory. These receptors have also been implicated in various neurodevelopmental disorders, such as Fragile X syndrome and autism. Internalization and recycling of these receptors in the neuron are important mechanisms to regulate the activity of the receptor and control the precise spatio-temporal localization of these receptors. Applying a "molecular replacement" approach in hippocampal neurons derived from mice, we demonstrate a critical role for protein interacting with C kinase 1 (PICK1) in regulating the agonist-induced internalization of mGluR1. We show that PICK1 specifically regulates the internalization of mGluR1 but it does not play any role in the internalization of the other member of group I mGluR family, mGluR5. Various regions of PICK1 viz., the N-terminal acidic motif, PDZ domain and BAR domain play important roles in the agonist-mediated internalization of mGluR1. Finally, we demonstrate that PICK1-mediated internalization of mGluR1 is critical for the resensitization of the receptor. Upon knockdown of endogenous PICK1, mGluR1s stayed on the cell membrane as inactive receptors, incapable of triggering the MAP-kinase signaling. They also could not induce AMPAR endocytosis, a cellular correlate for mGluR-dependent synaptic plasticity. Thus, this study unravels a novel role for PICK1 in the agonist-mediated internalization of mGluR1 and mGluR1-mediated AMPAR endocytosis that might contribute to the function of mGluR1 in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Namrata Ramsakha
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge city, Sector - 81, SAS Nagar, PO: 140306, Punjab, India
| | - Prachi Ojha
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge city, Sector - 81, SAS Nagar, PO: 140306, Punjab, India
| | - Subhajit Pal
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge city, Sector - 81, SAS Nagar, PO: 140306, Punjab, India
| | - Sanjeev Routh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge city, Sector - 81, SAS Nagar, PO: 140306, Punjab, India
| | - Ami Citri
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem; Edmond J. Safra Campus, Givat Ram, Jerusalem, Israel 91904; Institute of Life Sciences, The Hebrew University of Jerusalem; Edmond J. Safra Campus, Givat Ram, Jerusalem, Israel 91904; Program in Child and Brain Development, Canadian Institute for Advanced Research; MaRS Centre, West Tower, 661 University Ave, Suite 505, Toronto, Ontario, Canada M5G 1M1
| | - Samarjit Bhattacharyya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge city, Sector - 81, SAS Nagar, PO: 140306, Punjab, India.
| |
Collapse
|
3
|
Yadav P, Podia M, Kumari SP, Mani I. Glutamate receptor endocytosis and signaling in neurological conditions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:167-207. [PMID: 36813358 DOI: 10.1016/bs.pmbts.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The non-essential amino acid glutamate acts as a major excitatory neurotransmitter and plays a significant role in the central nervous system (CNS). It binds with two different types of receptors, ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs), responsible for the postsynaptic excitation of neurons. They are important for memory, neural development and communication, and learning. Endocytosis and subcellular trafficking of the receptor are essential for the regulation of receptor expression on the cell membrane and excitation of the cells. The endocytosis and trafficking of the receptor are dependent on its type, ligand, agonist, and antagonist present. This chapter discusses the types of glutamate receptors, their subtypes, and the regulation of their internalization and trafficking. The roles of glutamate receptors in neurological diseases are also briefly discussed.
Collapse
Affiliation(s)
- Prerna Yadav
- Department of Microbiology, University of Delhi, New Delhi, India
| | - Mansi Podia
- Department of Microbiology, University of Delhi, New Delhi, India
| | - Shashi Prabha Kumari
- Department of Microbiology, Ram Lal Anand College, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
4
|
Su LD, Wang N, Han J, Shen Y. Group 1 Metabotropic Glutamate Receptors in Neurological and Psychiatric Diseases: Mechanisms and Prospective. Neuroscientist 2021; 28:453-468. [PMID: 34088252 PMCID: PMC9449437 DOI: 10.1177/10738584211021018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors
that are activated by glutamate in the central nervous system (CNS).
Basically, mGluRs contribute to fine-tuning of synaptic efficacy and
control the accuracy and sharpness of neurotransmission. Among eight
subtypes, mGluR1 and mGluR5 belong to group 1 (Gp1) family, and are
implicated in multiple CNS disorders, such as Alzheimer’s disease,
autism, Parkinson’s disease, and so on. In the present review, we
systematically discussed underlying mechanisms and prospective of Gp1
mGluRs in a group of neurological and psychiatric diseases, including
Alzheimer’s disease, Parkinson’s disease, autism spectrum disorder,
epilepsy, Huntington’s disease, intellectual disability, Down’s
syndrome, Rett syndrome, attention-deficit hyperactivity disorder,
addiction, anxiety, nociception, schizophrenia, and depression, in
order to provide more insights into the therapeutic potential of Gp1
mGluRs.
Collapse
Affiliation(s)
- Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Na Wang
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ying Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Pandey S, Ramsakha N, Sharma R, Gulia R, Ojha P, Lu W, Bhattacharyya S. The post-synaptic scaffolding protein tamalin regulates ligand-mediated trafficking of metabotropic glutamate receptors. J Biol Chem 2020; 295:8575-8588. [PMID: 32376687 DOI: 10.1074/jbc.ra119.011979] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 05/04/2020] [Indexed: 11/06/2022] Open
Abstract
Group I metabotropic glutamate receptors (mGluRs) play important roles in various neuronal functions and have also been implicated in multiple neuropsychiatric disorders like fragile X syndrome, autism, and others. mGluR trafficking not only plays important roles in controlling the spatiotemporal localization of these receptors in the cell but also regulates the activity of these receptors. Despite this obvious significance, the cellular machineries that control the trafficking of group I metabotropic glutamate receptors in the central nervous system have not been studied in detail. The post-synaptic scaffolding protein tamalin has been shown to interact with group I mGluRs and also with many other proteins involved in protein trafficking in neurons. Using a molecular replacement approach in mouse hippocampal neurons, we show here that tamalin plays a critical role in the ligand-dependent internalization of mGluR1 and mGluR5, members of the group I mGluR family. Specifically, knockdown of endogenous tamalin inhibited the ligand-dependent internalization of these two receptors. Both N-terminal and C-terminal regions of tamalin played critical roles in mGluR1 endocytosis. Furthermore, we found that tamalin regulates mGluR1 internalization by interacting with S-SCAM, a protein that has been implicated in vesicular trafficking. Finally, we demonstrate that tamalin plays a critical role in mGluR-mediated internalization of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, a process believed to be the cellular correlate for mGluR-dependent synaptic plasticity. Taken together, these findings reveal a mechanistic role of tamalin in the trafficking of group I mGluRs and suggest its physiological implications in the brain.
Collapse
Affiliation(s)
- Saurabh Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, SAS Nagar, Punjab, India
| | - Namrata Ramsakha
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, SAS Nagar, Punjab, India
| | - Rohan Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, SAS Nagar, Punjab, India
| | - Ravinder Gulia
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, SAS Nagar, Punjab, India
| | - Prachi Ojha
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, SAS Nagar, Punjab, India
| | - Wei Lu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Samarjit Bhattacharyya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, SAS Nagar, Punjab, India
| |
Collapse
|
6
|
Hellyer SD, Albold S, Sengmany K, Singh J, Leach K, Gregory KJ. Metabotropic glutamate receptor 5 (mGlu 5 )-positive allosteric modulators differentially induce or potentiate desensitization of mGlu 5 signaling in recombinant cells and neurons. J Neurochem 2019; 151:301-315. [PMID: 31376155 DOI: 10.1111/jnc.14844] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/17/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022]
Abstract
Allosteric modulators of metabotropic glutamate receptor 5 (mGlu5 ) are a promising therapeutic strategy for a number of neurological disorders. Multiple mGlu5 -positive allosteric modulator (PAM) chemotypes have been discovered that act as either pure PAMs or as PAM-agonists in recombinant and native cells. While these compounds have been tested in paradigms of receptor activation, their effects on receptor regulatory processes are largely unknown. In this study, acute desensitization of mGlu5 mediated intracellular calcium mobilization by structurally diverse mGlu5 orthosteric and allosteric ligands was assessed in human embryonic kidney 293 cells and primary murine neuronal cultures from both striatum and cortex. We aimed to determine the intrinsic efficacy and modulatory capacity of diverse mGlu5 PAMs [(R)-5-((3-fluorophenyl)ethynyl)-N-(3-hydroxy-3-methylbutan-2-yl)picolinamide (VU0424465), N-cyclobutyl-6-((3-fluorophenyl)ethynyl)picolinamide (VU0360172), 1-(4-(2,4-difluorophenyl)piperazin-1-yl)-2-((4-fluorobenzyl)oxy)ethanone (DPFE), ((4-fluorophenyl) (2-(phenoxymethyl)-6,7-dihydrooxazolo[5,4-c]pyridin-5(4H)-yl)methanone) (VU0409551), 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB)] on receptor desensitization and whether cellular context influences receptor regulatory processes. Only VU0424465 and VU0409551 induced desensitization alone in human embryonic kidney 293-mGlu5 cells, while all PAMs enhanced (S)-3,5-dihydroxyphenylglycine (DHPG)-induced desensitization. All mGlu5 PAMs induced receptor desensitization alone and enhanced DHPG-induced desensitization in striatal neurons. VU0424465 and VU0360172 were the only PAMs that induced desensitization alone in cortical neurons. With the exception of (CDPPB), PAMs enhanced DHPG-induced desensitization in cortical neurons. Moreover, differential apparent affinities, efficacies, and cooperativities with DHPG were observed for VU0360172, VU0409551, and VU0424465 when comparing receptor activation and desensitization in a cell type-dependent manner. These data indicate that biased mGlu5 allosteric modulator pharmacology extends to receptor regulatory processes in a tissue dependent manner, adding yet another layer of complexity to rational mGlu5 drug discovery.
Collapse
Affiliation(s)
- Shane D Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Sabine Albold
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Kathy Sengmany
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Junaid Singh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
7
|
A Critical Role for Sorting Nexin 1 in the Trafficking of Metabotropic Glutamate Receptors. J Neurosci 2018; 38:8605-8620. [PMID: 30143569 DOI: 10.1523/jneurosci.0454-18.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/22/2018] [Accepted: 08/16/2018] [Indexed: 11/21/2022] Open
Abstract
Group I metabotropic glutamate receptors (mGluRs) function as modulators of neuronal physiology and they have also been implicated in various neuropsychiatric disorders. Trafficking of mGluRs plays important roles in controlling the precise localization of these receptors at specific region of the cell, as well as it regulates the activity of these receptors. Despite this obvious significance, we know very little about the cellular machineries that control the trafficking of these receptors in the CNS. Sorting nexin 1 (SNX1) has been shown to regulate the endosomal sorting of few cell surface receptors either to lysosomes where they are downregulated or back to the cell surface. Using "molecular replacement" approach in hippocampal neurons derived from mice of both sexes, we show here that SNX1 plays critical role in the trafficking of mGluR1, a member of the group I mGluR family. Overexpression of dominant-negative SNX1 or knockdown of endogenous SNX1 resulted in the rapid recycling of the receptor. Importantly, recycling via the rapid recycling route, did not allow the resensitization of the receptors. Our data suggest that both, N-terminal and C-terminal region of SNX1 play critical role in the normal trafficking of the receptor. In addition, we also show here that SNX1 regulates the trafficking of mGluR1 through the interaction with Hrs (hepatocyte growth factor-regulated tyrosine kinase substrate), a protein that has been implicated in both signaling and vesicular trafficking. Thus, these studies reveal a mechanistic role of SNX1 in the trafficking of group I mGluRs and its physiological implications.SIGNIFICANCE STATEMENT Group I mGluRs are activated by the neurotransmitter glutamate in the CNS, and play various important roles in the brain. Similar to many other receptors, trafficking plays crucial roles in controlling the precise localization as well as activity of these receptors. Despite this obvious significance very little is known about the cellular machineries that control the trafficking of these receptors. We demonstrate here, that SNX1 plays a critical role in the trafficking of mGluR1, a member of the group I mGluR family. SNX1-mediated trafficking is critical for the resensitization of the receptor. SNX1 controls the trafficking of the receptor through the interaction with another protein, Hrs. The results suggest a role for SNX1 in the regulation of group I mGluRs.
Collapse
|
8
|
Vergouts M, Doyen PJ, Peeters M, Opsomer R, Michiels T, Hermans E. PKC epsilon-dependent calcium oscillations associated with metabotropic glutamate receptor 5 prevent agonist-mediated receptor desensitization in astrocytes. J Neurochem 2017; 141:387-399. [PMID: 28266711 DOI: 10.1111/jnc.14007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/26/2017] [Accepted: 02/20/2017] [Indexed: 12/16/2022]
Abstract
A critical role has been assigned to protein kinase C (PKC)ε in the control of intracellular calcium oscillations triggered upon activation of type 5 metabotropic glutamate receptor (mGluR5) in cultured astrocytes. Nevertheless, the physiological significance of this particular signalling profile in the response of astrocytes to glutamate remains largely unknown. Considering that kinases are frequently involved in the regulation of G protein-coupled receptors, we have examined a putative link between the nature of the calcium signals and the response regulation upon repeated exposures of astrocytes to the agonist (S)-3,5-dihydroxyphenylglycine. We show that upon repeated mGluR5 activations, a robust desensitization was observed in astrocytes grown in culture conditions favouring the peak-plateau-type response. At variance, in cell cultures where calcium oscillations were predominating, the response was fully preserved even during repeated challenges with the agonist. Pharmacological inhibition of PKCε or genetic suppression of this isoform using shRNA was found to convert an oscillatory calcium profile to a sustained calcium mobilization and this latter profile was subject to desensitization upon repetitive mGluR5 activation. Our results suggest a yet undocumented scheme in which the activity of PKCε contributes to preserve the receptor sensitivity upon repeated or sustained activations. Cover Image for this issue: doi: 10.1111/jnc.13797.
Collapse
Affiliation(s)
- Maxime Vergouts
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, Brussels, Belgium
| | - Pierre J Doyen
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, Brussels, Belgium
| | - Michael Peeters
- Laboratory of virology, De Duve Institute, Université catholique de Louvain, Avenue Hippocrate B1.74.07, Brussels, Belgium
| | - Remi Opsomer
- Alzheimer Dementia Group, Institute of Neuroscience, Université catholique de Louvain, Avenue Mounier B1.53.02, Brussels, Belgium
| | - Thomas Michiels
- Laboratory of virology, De Duve Institute, Université catholique de Louvain, Avenue Hippocrate B1.74.07, Brussels, Belgium
| | - Emmanuel Hermans
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, Brussels, Belgium
| |
Collapse
|
9
|
Slifstein M, Abi-Dargham A. Recent Developments in Molecular Brain Imaging of Neuropsychiatric Disorders. Semin Nucl Med 2016; 47:54-63. [PMID: 27987558 DOI: 10.1053/j.semnuclmed.2016.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Molecular imaging with PET or SPECT has been an important research tool in psychiatry for as long as these modalities have been available. Here, we discuss two areas of neuroimaging relevant to current psychiatry research. The first is the use of imaging to study neurotransmission. We discuss the use of pharmacologic probes to induce changes in levels of neurotransmitters that can be inferred through their effects on outcome measures of imaging experiments, from their historical origins focusing on dopamine transmission through recent developments involving serotonin, GABA, and glutamate. Next, we examine imaging of neuroinflammation in the context of psychiatry. Imaging markers of neuroinflammation have been studied extensively in other areas of brain research, but they have more recently attracted interest in psychiatry research, based on accumulating evidence that there may be an inflammatory component to some psychiatric conditions. Furthermore, new probes are under development that would allow unprecedented insights into cellular processes. In summary, molecular imaging would continue to offer great potential as a unique tool to further our understanding of brain function in health and disease.
Collapse
Affiliation(s)
- Mark Slifstein
- Department of Psychiatry, Columbia University Medical Center, New York, NY; New York State Psychiatric Institute, New York, NY; Department of Psychiatry, Stony Brook University, New York, NY.
| | - Anissa Abi-Dargham
- Department of Psychiatry, Columbia University Medical Center, New York, NY; Department of Radiology, Columbia University Medical Center, New York, NY; New York State Psychiatric Institute, New York, NY; Department of Psychiatry, Stony Brook University, New York, NY
| |
Collapse
|
10
|
Raka F, Di Sebastiano AR, Kulhawy SC, Ribeiro FM, Godin CM, Caetano FA, Angers S, Ferguson SSG. Ca(2+)/calmodulin-dependent protein kinase II interacts with group I metabotropic glutamate and facilitates receptor endocytosis and ERK1/2 signaling: role of β-amyloid. Mol Brain 2015; 8:21. [PMID: 25885040 PMCID: PMC4378271 DOI: 10.1186/s13041-015-0111-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Agonist stimulation of Group I metabotropic glutamate receptors (mGluRs) initiates their coupling to the heterotrimeric G protein, Gαq/11, resulting in the activation of phospholipase C, the release of Ca(2+) from intracellular stores and the subsequent activation of protein kinase C. However, it is now recognized that mGluR5a also functions as a receptor for cellular prion protein (PrP(C)) and β-amyloid peptide (Aβ42) oligomers to facilitate intracellular signaling via the resulting protein complex. Intracellular mGluR5a signaling is also regulated by its association with a wide variety of intracellular regulation proteins. RESULTS In the present study, we utilized mass spectroscopy to identify calmodulin kinase IIα (CaMKIIα) as a protein that interacts with the second intracellular loop domain of mGluR5. We show that CaMKIIα interacts with both mGluR1a and mGluR5a in an agonist-independent manner and is co-immunoprecipitated with mGluR5a from hippocampal mouse brain. CaMKIIα positively regulates both mGluR1a and mGluR5a endocytosis, but selectively attenuates mGluR5a but not mGluR1a-stimulated ERK1/2 phosphorylation in a kinase activity-dependent manner. We also find that Aβ42 oligomers stimulate the association of CaMKIIα with mGluR5a and activate ERK1/2 in an mGluR5a-dependent manner. However, Aβ42 oligomer-stimulated ERK1/2 phosphorylation is not regulated by mGluR5a/CaMKIIα interactions suggesting that agonist and Aβ42 oligomers stabilize distinct mGluR5a activation states that are differentially regulated by CaMKIIα. The expression of both mGluR5a and PrP(C) together, but not alone resulted in the agonist-stimulated subcellular distribution of CaMKIIα into cytoplasmic puncta. CONCLUSIONS Taken together these results indicate that CaMKIIα selectively regulates mGluR1a and mGluR5a ERK1/2 signaling. As mGluR5 and CaMKIIα are involved in learning and memory and Aβ and mGluR5 are implicated in Alzheimer's disease, results of these studies could provide insight into potential pharmacological targets for treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Fitore Raka
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Andrea R Di Sebastiano
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Stephanie C Kulhawy
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Fabiola M Ribeiro
- Departamento de Bioquimica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - Christina M Godin
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Fabiana A Caetano
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Stephane Angers
- Leslie Dan Faculty of Pharmacy, University of Toronto, Room 901 144 College Street, Toronto, Ontario, Canada.
| | - Stephen S G Ferguson
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| |
Collapse
|
11
|
Nycthemeral variation in melatonin receptor expression in the lymphoid organs of a tropical seasonal breeder Funambulus pennanti. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2014; 200:1045-55. [DOI: 10.1007/s00359-014-0959-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 10/24/2014] [Accepted: 10/25/2014] [Indexed: 12/15/2022]
|
12
|
Pandey S, Mahato PK, Bhattacharyya S. Metabotropic glutamate receptor 1 recycles to the cell surface in protein phosphatase 2A-dependent manner in non-neuronal and neuronal cell lines. J Neurochem 2014; 131:602-14. [DOI: 10.1111/jnc.12930] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/30/2014] [Accepted: 08/08/2014] [Indexed: 02/03/2023]
Affiliation(s)
- Saurabh Pandey
- Department of Biological Sciences; Indian Institute of Science Education and Research (IISER) Mohali; Punjab India
| | - Prabhat Kumar Mahato
- Department of Biological Sciences; Indian Institute of Science Education and Research (IISER) Mohali; Punjab India
| | - Samarjit Bhattacharyya
- Department of Biological Sciences; Indian Institute of Science Education and Research (IISER) Mohali; Punjab India
| |
Collapse
|
13
|
Rab8 modulates metabotropic glutamate receptor subtype 1 intracellular trafficking and signaling in a protein kinase C-dependent manner. J Neurosci 2013; 32:16933-42a. [PMID: 23175844 DOI: 10.1523/jneurosci.0625-12.2012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs) are G protein-coupled receptors (GPCRs) that are activated by glutamate, the primary excitatory neurotransmitter in the CNS. Alterations in glutamate receptor signaling are implicated in neuropathologies such as Alzheimer's disease, ischemia, and Huntington's disease among others. Group 1 mGluRs (mGluR1 and mGluR5) are primarily coupled to Gα(q/11) leading to the activation of phospholipase C and the formation of diacylglycerol and inositol 1,4,5-trisphosphate, which results in the release of intracellular calcium stores and protein kinase C (PKC) activation. Desensitization, endocytosis, and recycling are major mechanisms of GPCR regulation, and the intracellular trafficking of GPCRs is linked to the Rab family of small G proteins. Rab8 is a small GTPase that is specifically involved in the regulation of secretory/recycling vesicles, modulation of the actin cytoskeleton, and cell polarity. Rab8 has been shown to regulate the synaptic delivery of AMPA receptors during long-term potentiation and during constitutive receptor recycling. We show here that Rab8 interacts with the C-terminal tail of mGluR1a in an agonist-dependent manner and plays a role in regulating of mGluR1a signaling and intracellular trafficking in human embryonic kidney 293 cells. Specifically, Rab8 expression attenuates mGluR1a-mediated inositol phosphate formation and calcium release from mouse neurons in a PKC-dependent manner, while increasing cell surface mGluR1a expression via decreased receptor endocytosis. These experiments provide us with an understanding of the role Rabs play in coordinated regulation of mGluR1a and how this impacts mGluR1a signaling.
Collapse
|
14
|
Bisgaard CF, Bak S, Christensen T, Jensen ON, Enghild JJ, Wiborg O. Vesicular signalling and immune modulation as hedonic fingerprints: proteomic profiling in the chronic mild stress depression model. J Psychopharmacol 2012; 26:1569-83. [PMID: 23139383 DOI: 10.1177/0269881112460110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Extensive preclinical research has focused at unravelling the underlying molecular mechanisms leading to depression and recovery. In this study, we investigated the quantitative changes in protein abundance in the ventral hippocampal granular cell layer. We compared different phenotypes from the chronic mild stress (CMS) model of depression using chronic administration with two selective serotonin reuptake inhibitors (SSRIs), escitalopram and sertraline. We isolated granular cells using Laser-Capture Microdissection (LCM) and we identified their regulated proteins using two-dimensional (2D) differential gel electrophoresis (DIGE) and tandem mass spectrometry (MS/MS). The majority of the proteins we identified were enzymes involved in different metabolic activities. Additional proteins were functionally classified as vesicular proteins and immune system proteins. Rab GDP dissociation inhibitor alpha (GDIA) and syntaxin-binding protein 1 (STXB1) were potential markers for stress reactivity. Dynamin 1 (DYN1), glutathione S-transferase omega-1 (GSTO1) and peroxiredoxin (PRDX6) were associated with treatment response. In addition, an imbalance between different post-translationally modified versions of DYN1 and GSTO1 potentially accounted for SSRI treatment refraction. In the present study, we searched for new markers of stress reactivity and treatment response as well as any underlying molecular mechanisms correlating to the development of anhedonia and antidepressant therapy refraction. Our results pointed towards an essential role of post-translational modifications in both vesicular and immune protein systems.
Collapse
Affiliation(s)
- Christina F Bisgaard
- Centre for Psychiatric Research, Institute of Clinical Medicine, Aarhus University Hospital, Risskov, Denmark.
| | | | | | | | | | | |
Collapse
|
15
|
Foss CA, Mease RC, Cho SY, Kim HJ, Pomper MG. GCPII imaging and cancer. Curr Med Chem 2012; 19:1346-59. [PMID: 22304713 DOI: 10.2174/092986712799462612] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 12/26/2011] [Accepted: 12/27/2011] [Indexed: 12/11/2022]
Abstract
Glutamate carboxypeptidase II (GCPII) in the central nervous system is referred to as the prostate-specific membrane antigen (PSMA) in the periphery. PSMA serves as a target for imaging and treatment of prostate cancer and because of its expression in solid tumor neovasculature has the potential to be used in this regard for other malignancies as well. An overview of GCPII/PSMA in cancer, as well as a discussion of imaging and therapy of prostate cancer using a wide variety of PSMA-targeting agents is provided.
Collapse
Affiliation(s)
- C A Foss
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical School, Baltimore, MD 21231, USA
| | | | | | | | | |
Collapse
|
16
|
p38 mitogen-activated protein kinase inhibitor reduces neurocan production in cultured spinal cord astrocytes. Neuroreport 2012; 23:546-50. [DOI: 10.1097/wnr.0b013e328354256c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
17
|
Martins-de-Souza D. Proteomics as a tool for understanding schizophrenia. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2011; 9:95-101. [PMID: 23430140 PMCID: PMC3569116 DOI: 10.9758/cpn.2011.9.3.95] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 07/01/2011] [Accepted: 07/06/2011] [Indexed: 11/18/2022]
Abstract
Schizophrenia is likely to be a multifactorial disorder, consequence of alterations in gene and protein expression since the neurodevelopment that together to environmental factors will trigger the establishment of the disease. In the post-genomic era, proteomics has emerged as a promising strategy for revealing disease and treatment biomarkers as well as a tool for the comprehension of the mechanisms of schizophrenia pathobiology. Here, there is a discussion of the potential pathways and structures that are compromised in schizophrenia according to proteomic findings while studying five distinct brain regions of post-mortem tissue from schizophrenia patients and controls. Proteins involved in energy metabolism, calcium homeostasis, myelinization, and cytoskeleton have been recurrently found to be differentially expressed in schizophrenia brains. These findings may encourage new studies on the understanding of schizophrenia biochemical pathways and even new potential drug targets.
Collapse
Affiliation(s)
- Daniel Martins-de-Souza
- Max Planck Institute of Psychiatry, Munich, Germany and Lab. de Neurociências (LIM-27), Inst. Psiquiatria, Fac. de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Price DL, Rockenstein E, Ubhi K, Phung V, MacLean-Lewis N, Askay D, Cartier A, Spencer B, Patrick C, Desplats P, Ellisman MH, Masliah E. Alterations in mGluR5 expression and signaling in Lewy body disease and in transgenic models of alpha-synucleinopathy--implications for excitotoxicity. PLoS One 2010; 5:e14020. [PMID: 21103359 PMCID: PMC2982819 DOI: 10.1371/journal.pone.0014020] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Accepted: 10/19/2010] [Indexed: 12/21/2022] Open
Abstract
Dementia with Lewy bodies (DLB) and Parkinson's Disease (PD) are neurodegenerative disorders of the aging population characterized by the abnormal accumulation of alpha-synuclein (alpha-syn). Previous studies have suggested that excitotoxicity may contribute to neurodegeneration in these disorders, however the underlying mechanisms and their relationship to alpha-syn remain unclear. For this study we proposed that accumulation of alpha-syn might result in alterations in metabotropic glutamate receptors (mGluR), particularly mGluR5 which has been linked to deficits in murine models of PD. In this context, levels of mGluR5 were analyzed in the brains of PD and DLB human cases and alpha-syn transgenic (tg) mice and compared to age-matched, unimpaired controls, we report a 40% increase in the levels of mGluR5 and beta-arrestin immunoreactivity in the frontal cortex, hippocampus and putamen in DLB cases and in the putamen in PD cases. In the hippocampus, mGluR5 was more abundant in the CA3 region and co-localized with alpha-syn aggregates. Similarly, in the hippocampus and basal ganglia of alpha-syn tg mice, levels of mGluR5 were increased and mGluR5 and alpha-syn were co-localized and co-immunoprecipitated, suggesting that alpha-syn interferes with mGluR5 trafficking. The increased levels of mGluR5 were accompanied by a concomitant increase in the activation of downstream signaling components including ERK, Elk-1 and CREB. Consistent with the increased accumulation of alpha-syn and alterations in mGluR5 in cognitive- and motor-associated brain regions, these mice displayed impaired performance in the water maze and pole test, these behavioral alterations were reversed with the mGluR5 antagonist, MPEP. Taken together the results from study suggest that mGluR5 may directly interact with alpha-syn resulting in its over activation and that this over activation may contribute to excitotoxic cell death in select neuronal regions. These results highlight the therapeutic importance of mGluR5 antagonists in alpha-synucleinopathies.
Collapse
Affiliation(s)
- Diana L. Price
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
| | - Edward Rockenstein
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Kiren Ubhi
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Van Phung
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
- Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - Natalie MacLean-Lewis
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
- Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - David Askay
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
| | - Anna Cartier
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Brian Spencer
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Christina Patrick
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Paula Desplats
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Mark H. Ellisman
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California, United States of America
- Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - Eliezer Masliah
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Department of Pathology, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
19
|
Martins-De-Souza D, Dias-Neto E, Schmitt A, Falkai P, Gormanns P, Maccarrone G, Turck CW, Gattaz WF. Proteome analysis of schizophrenia brain tissue. World J Biol Psychiatry 2010; 11:110-20. [PMID: 20109112 DOI: 10.3109/15622970903490626] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Proteome analysis has emerged as a promising strategy to the identification of potential biomarkers and to further confirm the importance of certain pathways in the schizophrenia (SCZ) pathophysiology. Reviewing the results of 13 proteome studies in SCZ brain tissue, we aimed to provide information regarding potential proteins biomarkers as well as information about the pathophysiology of the disease. METHODS AND RESULTS Using two-dimensional gel electrophoresis and shotgun mass spectrometry, 31 proteins were consistently found differentially expressed in the brains of SCZ patients. The most frequent protein alterations reported in SCZ were related to brain energy metabolism, brain plasticity, and synaptic function, processes that are thought to belong to the core of the biology of this disease. The recurrent identification and validation of inter-related protein clusters, determined in different samples and approaches, strongly reinforces the putative involvement of certain pathways in SCZ. CONCLUSIONS The availability of reliable markers not only paves the way to the development of new therapeutic strategies but also points out the possibility of their use as peripheral blood markers that may potentially contribute to the early SCZ detection and early therapeutic intervention, both of which can reduce the social and cognitive consequences of the disease.
Collapse
|
20
|
Kang M, Othmer HG. Spatiotemporal characteristics of calcium dynamics in astrocytes. CHAOS (WOODBURY, N.Y.) 2009; 19:037116. [PMID: 19792041 PMCID: PMC2852438 DOI: 10.1063/1.3206698] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2009] [Accepted: 07/24/2009] [Indexed: 05/28/2023]
Abstract
Although Ca(i)(2+) waves in networks of astrocytes in vivo are well documented, propagation in vivo is much more complex than in culture, and there is no consensus concerning the dominant roles of intercellular and extracellular messengers [inositol 1,4,5-trisphosphate (IP(3)) and adenosine-5'-triphosphate (ATP)] that mediate Ca(i)(2+) waves. Moreover, to date only simplified models that take very little account of the geometrical struture of the networks have been studied. Our aim in this paper is to develop a mathematical model based on realistic cellular morphology and network connectivity, and a computational framework for simulating the model, in order to address these issues. In the model, Ca(i) (2+) wave propagation through a network of astrocytes is driven by IP(3) diffusion between cells and ATP transport in the extracellular space. Numerical simulations of the model show that different kinetic and geometric assumptions give rise to differences in Ca(i)(2+) wave propagation patterns, as characterized by the velocity, propagation distance, time delay in propagation from one cell to another, and the evolution of Ca(2+) response patterns. The temporal Ca(i)(2+) response patterns in cells are different from one cell to another, and the Ca(i)(2+) response patterns evolve from one type to another as a Ca(i)(2+) wave propagates. In addition, the spatial patterns of Ca(i)(2+) wave propagation depend on whether IP(3), ATP, or both are mediating messengers. Finally, two different geometries that reflect the in vivo and in vitro configuration of astrocytic networks also yield distinct intracellular and extracellular kinetic patterns. The simulation results as well as the linear stability analysis of the model lead to the conclusion that Ca(i)(2+) waves in astrocyte networks are probably mediated by both intercellular IP(3) transport and nonregenerative (only the glutamate-stimulated cell releases ATP) or partially regenerative extracellular ATP signaling.
Collapse
Affiliation(s)
- Minchul Kang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
21
|
Bidirectional Hebbian plasticity at hippocampal mossy fiber synapses on CA3 interneurons. J Neurosci 2009; 28:14042-55. [PMID: 19109487 DOI: 10.1523/jneurosci.4848-08.2008] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hippocampal area CA3 is critically involved in the formation of nonoverlapping neuronal subpopulations ("pattern separation") to store memory representations as distinct events. Efficient pattern separation relies on the strong and sparse excitatory input from the mossy fibers (MFs) to pyramidal cells and feedforward inhibitory interneurons. However, MF synapses on CA3 pyramidal cells undergo long-term potentiation (LTP), which, if unopposed, will degrade pattern separation because MF activation will now recruit additional CA3 pyramidal cells. Here, we demonstrate MF LTP in stratum lacunosum-moleculare (L-M) interneurons induced by the same stimulation protocol that induces MF LTP in pyramidal cells. This LTP was NMDA receptor (NMDAR) independent and occurred at MF Ca(2+)-impermeable AMPA receptor synapses. LTP was prevented by with voltage clamping the postsynaptic cell soma during high-frequency stimulation (HFS), intracellular injections of the Ca(2+) chelator BAPTA (20 mm), or bath applications of the L-type Ca(2+) channel blocker nimodipine (10 microm). We propose that MF LTP in L-M interneurons preserves the sparsity of pyramidal cell activation, thus allowing CA3 to maintain its role in pattern separation. In the presence of the mGluR1alpha antagonist LY367385 [(S)-(+)-a-amino-4-carboxy-2-methylbenzeneacetic acid] (100 microm), the same HFS that induces MF LTP in naive slices triggered NMDAR-independent MF LTD. This LTD, like LTP, required activation of the L-type Ca(2+) channel and also was induced after blockade of IP(3) receptors with heparin (4 mg/ml) or the selective depletion of receptor-gated Ca(2+) stores with ryanodine (10 or 100 microm). We conclude that L-M interneurons are endowed with Ca(2+) signaling cascades suitable for controlling the polarity of MF long-term plasticity induced by joint presynaptic and postsynaptic activities.
Collapse
|
22
|
Ferraguti F, Crepaldi L, Nicoletti F. Metabotropic glutamate 1 receptor: current concepts and perspectives. Pharmacol Rev 2008; 60:536-81. [PMID: 19112153 DOI: 10.1124/pr.108.000166] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Almost 25 years after the first report that glutamate can activate receptors coupled to heterotrimeric G-proteins, tremendous progress has been made in the field of metabotropic glutamate receptors. Now, eight members of this family of glutamate receptors, encoded by eight different genes that share distinctive structural features have been identified. The first cloned receptor, the metabotropic glutamate (mGlu) receptor mGlu1 has probably been the most extensively studied mGlu receptor, and in many respects it represents a prototypical subtype for this family of receptors. Its biochemical, anatomical, physiological, and pharmacological characteristics have been intensely investigated. Together with subtype 5, mGlu1 receptors constitute a subgroup of receptors that couple to phospholipase C and mobilize Ca(2+) from intracellular stores. Several alternatively spliced variants of mGlu1 receptors, which differ primarily in the length of their C-terminal domain and anatomical localization, have been reported. Use of a number of genetic approaches and the recent development of selective antagonists have provided a means for clarifying the role played by this receptor in a number of neuronal systems. In this article we discuss recent advancements in the pharmacology and concepts about the intracellular transduction and pathophysiological role of mGlu1 receptors and review earlier data in view of these novel findings. The impact that this new and better understanding of the specific role of these receptors may have on novel treatment strategies for a variety of neurological and psychiatric disorders is considered.
Collapse
Affiliation(s)
- Francesco Ferraguti
- Department of Pharmacology, Innsbruck Medical University, Peter-Mayr Strasse 1a, Innsbruck A-6020, Austria.
| | | | | |
Collapse
|
23
|
Effects of subtype-selective group I mGluR antagonists on synchronous activity induced by 4-aminopyridine/CGP 55845 in adult guinea pig hippocampal slices. Neuropharmacology 2008; 55:47-54. [PMID: 18538357 DOI: 10.1016/j.neuropharm.2008.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2008] [Revised: 04/14/2008] [Accepted: 04/16/2008] [Indexed: 12/26/2022]
Abstract
Co-application of the convulsant 4-aminopyridine (4-AP) and the GABA(B) receptor antagonist CGP 55845 to adult guinea pig hippocampal slices elicits giant GABA-mediated postsynaptic potentials (GPSPs) and epileptiform discharges. Here we tested the effects of the group I metabotropic glutamate receptor (mGluR) subtype-selective antagonists LY 367385 (mGlu1, 100 microM), MPEP (mGlu5, 10 microM), and MTEP (mGlu5, 500 nM) on this synchronous activity. Electrophysiological field recordings were performed in the CA3 region of hippocampal slices from adult guinea pigs. The mGlu5 receptor antagonists increased GPSP rate, but the mGlu1 receptor antagonist did not. This ability of mGlu5 receptor antagonists to increase the rate of GPSPs indicates that enough endogenous glutamate is released under these conditions to activate group I mGluR; nevertheless, co-application of a mGlu1 receptor antagonist (LY 367385 or JNJ 16259685) and MPEP did not decrease pre-existing epileptiform activity. Furthermore, co-application of LY 367,385 and MPEP did not prevent the emergence of epileptiform activity. When ionotropic glutamate receptor (iGluR) antagonists were present, neither MPEP nor the group I mGluR agonist DHPG changed GPSP rate, suggesting that pyramidal cell-to-interneuron iGluR-mediated synaptic connections are involved in the rate change mechanism. In contrast to the lack of effect of group I mGluR antagonists on epileptiform activity in the 4-AP/CGP 55845 model, group I mGluR antagonists blocked the emergence of longer epileptiform events and decreased the overall amount of synchronous activity in the GABA(A) antagonist/4-AP model. In conclusion, in the 4-AP/CGP 55845 model, enough glutamate was released to activate group I mGluRs and affect GPSP rate via mGlu5 receptors; however, this group I mGluR activation was not required for the generation of the epileptiform activity.
Collapse
|
24
|
Abstract
Synaptic transmission from photoreceptors to all types of ON bipolar cells is primarily mediated by the mGluR6 receptor. This receptor, which is apparently expressed uniquely in the nervous system by ON bipolar cells, couples negatively to a nonselective cation channel. This arrangement results in a sign reversal at photoreceptor/ON bipolar cell synapse, which is necessary in order to establish parallel ON and OFF pathways in the retina. The synapse is an important target for second messenger molecules that are known to modulate synaptic transmission elsewhere in the nervous system, second messengers that act on a time scale ranging from milliseconds to minutes. This review focuses on two of these molecules, Ca2+ and cGMP, summarizing our current knowledge of how they modulate gain at the photoreceptor/ON bipolar cell synapse, as well as their proposed sites of action within the mGluR6 cascade. The implications of plasticity at this synapse for retinal function will also be examined.
Collapse
Affiliation(s)
- Josefin Snellman
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, SHM-B103, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
25
|
Werner CG, Scartabelli T, Pancani T, Landucci E, Moroni F, Pellegrini-Giampietro DE. Differential role of mGlu1 and mGlu5 receptors in rat hippocampal slice models of ischemic tolerance. Eur J Neurosci 2007; 25:3597-604. [PMID: 17610579 DOI: 10.1111/j.1460-9568.2007.05614.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Activation of glutamate receptors has been proposed as a key factor in the induction of ischemic tolerance. We used organotypic rat hippocampal slices exposed to 30 min oxygen-glucose deprivation (OGD) to evaluate postischemic pyramidal cell death in the CA1 subregion. In this model, 10 min exposure to OGD 24 h before the exposure to toxic OGD was not lethal and reduced the subsequent OGD neurotoxicity by approximately 53% (ischemic preconditioning). Similarly, a 30 min exposure to the group I mGlu receptor agonist DHPG (10 microM) significantly reduced OGD neurotoxicity 24 h later (pharmacological preconditioning). Ischemic tolerance did not develop when either the selective mGlu1 antagonists LY367385 and 3-MATIDA or the AMPA/KA antagonist CNQX were present in the incubation medium during exposure to sublethal OGD. Neither the NMDA antagonist MK801 nor the mGlu5 antagonist MPEP affected the preconditioning process. On the other hand, pharmacological preconditioning was prevented not only by LY367385 or CNQX, but also by MPEP. In preconditioned slices, the toxic responses to AMPA or NMDA were reduced. The neurotoxicty of 100 microM DHPG in slices simultaneously exposed to a mild (20 min) OGD was differentially altered in the two preconditioning paradigms. After ischemic preconditioning, DHPG neurotoxicity was reduced in a manner that was sensitive to LY367385 but not to MPEP, whereas after pharmacological preconditioning it was enhanced in a manner that was sensitive to MPEP but not to LY367385. Our results show that mGlu1 and mGlu5 receptors are differentially involved in the induction and expression of ischemic tolerance following two diverse preconditioning stimuli.
Collapse
Affiliation(s)
- Claudia G Werner
- Dipartimento di Farmacologia Preclinica e Clinica, Universitá di Firenze, Viale G. Pieraccini 6, 50139 Firenze, Italy
| | | | | | | | | | | |
Collapse
|
26
|
Mathiesen JM, Ramirez MT. The metabotropic glutamate receptor 4 is internalized and desensitized upon protein kinase C activation. Br J Pharmacol 2006; 148:279-90. [PMID: 16582932 PMCID: PMC1751557 DOI: 10.1038/sj.bjp.0706733] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. The metabotropic glutamate receptor 4 (mGluR4) is a Galphai-coupled receptor that modulates glutamatergic neurotransmission. As mGluR4 expression and activation have been implicated in a number of pathological conditions and because the internalization and desensitization properties of this receptor are poorly understood, studies were designed to investigate these aspects of mGluR4 biology. 2. Neither agonist activation by L-(+)-2-amino-4-phosphonobutyric acid (L-AP4) nor L-glutamate caused mGluR4 internalization when cmyc-tagged mGluR4 was expressed in a human embryonic kidney 293 cell line as assessed by cell surface enzyme-linked immunosorbent and immunostaining assays. Instead, a modest increase in mGluR4 surface expression was observed and found to be receptor specific as the competitive antagonist alpha-cyclopropyl-4-phosphonophenylglycine (CPPG) blocked this effect. 3. In contrast, mGluR4 internalized when the protein kinase C (PKC) pathway was activated either by phorbol-12-myristate-13-acetate (PMA) or by the activation of the Galphaq-coupled, neurokinin 3 receptor (NK3R) when co-expressed. This process was PKC-dependent as the specific PKC inhibitor GF 109203X inhibited PMA and NK3R-mediated internalization. 4. PKC activation by PMA caused desensitization of mGluR4 as measured by forskolin-stimulated cAMP inhibition, whereas agonist activation had no effect on desensitization. 5. When mGluR4's coupling was redirected from adenylyl cyclase to phospholipase C by coexpression of a chimeric Galphaqo5 protein, mGluR4 both internalized and desensitized in response to its agonists. 6. These findings demonstrate that mGluR4 internalization and desensitization are agonist-independent unless pathways leading to the activation of PKC are induced.
Collapse
Affiliation(s)
- Jesper Mosolff Mathiesen
- Department of Molecular Pharmacology, H. Lundbeck A/S, DK-2500 Valby, Denmark
- Department of Medicinal Chemistry, Danish University of Pharmaceutical Sciences, DK-2100 Copenhagen, Denmark
| | - M Teresa Ramirez
- Department of Molecular Pharmacology, H. Lundbeck A/S, DK-2500 Valby, Denmark
- Author for correspondence:
| |
Collapse
|
27
|
Watanabe S, Hong M, Lasser-Ross N, Ross WN. Modulation of calcium wave propagation in the dendrites and to the soma of rat hippocampal pyramidal neurons. J Physiol 2006; 575:455-68. [PMID: 16809362 PMCID: PMC1819440 DOI: 10.1113/jphysiol.2006.114231] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/25/2006] [Accepted: 06/26/2006] [Indexed: 01/03/2023] Open
Abstract
Repetitive synaptic stimulation in the stratum radiatum (SR) evokes large amplitude Ca2+ waves in the thick apical dendrites of hippocampal CA1 pyramidal neurons. These waves are initiated by activation of metabotropic glutamate receptors (mGluRs), which mobilize inositol-1,4,5-trisphospate (IP3) and release Ca2+ from intracellular stores. We explored mechanisms that modulate the spatial properties of these waves. Higher stimulus current evoked waves of increasing spatial extent. Most waves did not propagate through the soma; the majority stopped close to the junction of the soma and apical dendrite. Pairing strong stimulation with one electrode and subthreshold stimulation with another (associative activation) extended the waves distally but failed to extend waves into the cell body. Pairing synaptic stimulation with backpropagating action potentials enhanced the likelihood of wave generation but did not extend the waves to the somatic region. Priming the stores with Ca2+ entry through voltage dependent channels modulated wave properties but did not extend them past the dendrites. These results are consistent with propagation failing due to the dilution of synaptically generated IP3 as it diffuses into the large volume of the soma (impedance mismatch). Synaptically activating waves in the presence of low concentrations of carbachol, which probably increased the tonic level of IP3 throughout the cell, enhanced the extent of propagation and generated waves that invaded the soma, as long as low-affinity indicators were used to detect the [Ca2+]i changes. Consistent with this explanation direct injection of IP3 into the soma promoted wave propagation into this region. Ca2+ waves that propagated through the cell body were interesting because they did not fill the volume of the soma, but passed through the centre, often with large amplitude. These waves may be particularly effective in activating gene expression and protein synthesis.
Collapse
Affiliation(s)
- Shigeo Watanabe
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | |
Collapse
|
28
|
Nakamichi N, Yoneda Y. Maturation-dependent reduced responsiveness of intracellular free Ca2+ ions to repeated stimulation by N-methyl-d-aspartate in cultured rat cortical neurons. Neurochem Int 2006; 49:230-7. [PMID: 16517022 DOI: 10.1016/j.neuint.2006.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 01/10/2006] [Accepted: 01/17/2006] [Indexed: 11/25/2022]
Abstract
In contrast to other ionotropic glutamate receptors, N-methyl-d-aspartate (NMDA) receptor channels are rather stable after the simulation. Brief exposure to NMDA at 50 microM rapidly increased the fluorescence intensity for increased intracellular free Ca(2+) levels in a reversible- and concentration-dependent manner in rat cortical neurons cultured for 3-15 days in vitro (DIV), while EC(50) values were significantly decreased in proportion to cellular maturation from 3 to 15 DIV. Although a constant increase was persistently seen in the fluorescence throughout the sustained exposure to NMDA for 60 min irrespective of the cell maturation from 3 to 15 DIV, the second brief exposure for 5 min resulted in a less efficient increase in the fluorescence than that found after the first brief exposure for 5 min in a manner dependent on intervals between the two repetitive brief exposures. In vitro maturation significantly shortened the interval required for the reduced responsiveness to the second brief exposure, while in immature neurons prolonged intervals were required for the reduced responsiveness to the second brief exposure to NMDA. Moreover, brief exposure to NMDA led to a marked decrease in immunoreactivity to extracellular loop of NR1 subunit in cultured neurons not permeabilized in proportion to the time after washing. These results suggest that cellular maturation would facilitate the desensitization process to repeated stimulation by NMDA, without markedly affecting that to sustained stimulation, through a mechanism related to the decreased number of NMDA receptors expressed at cell surfaces in cultured rat cortical neurons.
Collapse
Affiliation(s)
- Noritaka Nakamichi
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Natural Science and Technology, Kakuma-machi, Ishikawa 920-1192, Japan
| | | |
Collapse
|
29
|
Do SH, Yun HS, Jeong WI, Jeong DH, Ki MR, Chung JY, Park SJ, Kim SB, Jeong KS. Up-regulation of Metabotropic glutamate receptor 3 (mGluR3) in rat fibrosis and cirrhosis model of persistent hypoxic condition. Mol Cell Biochem 2006; 294:189-96. [PMID: 16845489 DOI: 10.1007/s11010-006-9259-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2006] [Accepted: 06/01/2006] [Indexed: 10/24/2022]
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system, and evidence for peripheral glutamatergic fibers in mammals is still lacking. However, glutamate receptors have been identified in peripheral organs, including taste buds, myenteric plexus, and pancreatic islet cell. Protection against anoxic damage could also be explained by mechanisms mediated by postsynaptic mGluR2 or mGluR3, such as the inhibition of membrane excitability resulting from a reduction of cAMP formation by a G-protein-dependent modulation of ion channels. In addition, activation of mGluR3 present in glial cells may contribute to neuroprotection by enhancing the production of death. Thus, mGluR2/3 behaves potentially as a major defensive mechanism anoxia-tolerant species. There are a few reports for the regional pattern of hypoxic damage, which was inversely related to the expression of mGluR2/3. The aim of this study was to characterize the expression of mGluR3 in hypoxic liver in experimental model of rat liver. Proteomic analysis of protein extracts from CCl4-induced cirrhotic liver revealed the presence of the mGluR3. The presence of mGluR3 in the cirrhotic liver was confirmed by immunohistochemical analysis. There were a number of macrophages expressing mGluR3 mainly in the fibrous septa. After 2 weeks recovery, however, most of mGluR3 positive macrophages disappeared with collagen fibers. These results demonstrate that mGluR3 involved in the liver in response to persistent hypoxic status such as fibrotic/cirrhotic condition, and suggest that the expression of mGluR3 may be a key role functional metabolism and viability in the liver by interacting with the glutamate receptors in vivo.
Collapse
Affiliation(s)
- Sun Hee Do
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, 702-701, #1370 Sangyeok-dong, Buk-gu, Daegu, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Dhami GK, Ferguson SSG. Regulation of metabotropic glutamate receptor signaling, desensitization and endocytosis. Pharmacol Ther 2006; 111:260-71. [PMID: 16574233 DOI: 10.1016/j.pharmthera.2005.01.008] [Citation(s) in RCA: 155] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2005] [Accepted: 01/25/2005] [Indexed: 11/24/2022]
Abstract
Metabotropic glutamate receptors (mGluRs) comprise a unique family of G protein-coupled receptors (GPCR) that can be classified into 3 groups based on G protein coupling specificity and sequence similarity. Group I mGluRs (mGluR1 and mGluR5) are coupled to the heterotrimeric G protein Galpha(q/11) and trigger the release of calcium from intracellular stores. In the present review, we discuss the molecular mechanisms involved in the desensitization and endocytosis of group I mGluRs. Group I mGluRs desensitize in response to both second-messenger-dependent protein kinases and G protein-coupled receptor kinases (GRK). However, GRK2-mediated mGluR1 desensitization appears to be both phosphorylation- and beta-arrestin-independent. In addition to GRK-mediated uncoupling of mGluRs from heterotrimeric G proteins, the huntingtin-interacting protein, optineurin, also contributes to mGluR1 and mGluR5 desensitization. The G protein-uncoupling activity of optineurin appears to be facilitated by the presence of polyglutamine-expanded mutant huntingtin but not wild-type huntingtin. Group I mGluRs also undergo both agonist-dependent and -independent endocytosis in both heterologous cell expression systems and primary neuronal cultures. The present review overviews the current understanding of the contribution of second messenger-dependent protein kinases, beta-arrestins and a novel Ral/phospholipase D2 (PLD2)-mediated endocytic pathway to the regulation of Group I mGluR endocytosis. Overall, the regulation of Group I mGluR desensitization and endocytosis appears to be mediated by the same molecular intermediates as have been described for more typical GPCR such as the beta(2)-adrenergic receptor. However, there appears to be subtle, but important, differences in the mechanisms by which these intermediates are employed to regulate Group I mGluR desensitization and endocytosis.
Collapse
Affiliation(s)
- Gurpreet K Dhami
- Cell Biology Research Group, Robarts Research Institute, London, Ontario, Canada N6A 5K8
| | | |
Collapse
|
31
|
Mitrano DA, Smith Y. Comparative analysis of the subcellular and subsynaptic localization of mGluR1a and mGluR5 metabotropic glutamate receptors in the shell and core of the nucleus accumbens in rat and monkey. J Comp Neurol 2006; 500:788-806. [PMID: 17154259 DOI: 10.1002/cne.21214] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Group I metabotropic glutamate receptors (mGluRs) play critical roles in synaptic plasticity and drug addiction. To characterize potential sites whereby these receptors mediate their effects in the ventral striatum, we studied the subcellular and subsynaptic localization of mGluR1a and mGluR5 in the shell and core of the nucleus accumbens in rat and monkey. In both species, group I mGluRs are mainly postsynaptic in dendrites and spines, with rare presynaptic labeling in unmyelinated axons. Minor, yet significant, differences in proportions of specific immunoreactive elements were found between the accumbens shell and the accumbens core in monkey. At the subsynaptic level, significant differences were found in the proportion of plasma membrane-bound mGluR5 labeling between species. In dendrites, spines, and unmyelinated axons, a significantly larger proportion of mGluR5 labeling was bound to the plasma membrane in rats (50-70%) than in monkeys (30-50%). Conversely, mGluR1a displayed the same pattern of immunogold labeling in the two species. Electron microscopic colocalization studies revealed 30% colocalization of mGluR1a and mGluR5 in dendrites and as much as 50-65% in spines in both compartments of the rat accumbens. Both group I mGluRs were significantly expressed in D1-immunoreactive dendritic processes (60-75% colocalization) and spines (30-50%) of striatal projection neurons as well as dendrites of cholinergic (30-70%) and parvalbumin-containing (70-85%) interneurons. These findings highlight the widespread expression of group I mGluRs in projection neurons and interneurons of the shell and core of the nucleus accumbens, providing a solid foundation for regulatory and therapeutic functions of group I mGluRs in reward-related behaviors and drug addiction.
Collapse
Affiliation(s)
- Darlene A Mitrano
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
32
|
Chang HJ, Yoo BC, Lim SB, Jeong SY, Kim WH, Park JG. Metabotropic glutamate receptor 4 expression in colorectal carcinoma and its prognostic significance. Clin Cancer Res 2005; 11:3288-95. [PMID: 15867225 DOI: 10.1158/1078-0432.ccr-04-1912] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Metabotropic glutamate receptors (mGluR) play a variety of roles in both neuronal and nonneuronal cells. Recently, we reported that mGluR4 mediates 5-fluorouracil resistance in a human colon cancer cell line. In this study, we evaluated the nonneural expression of mGluR4 and clarified the existence of mGluR4 in normal colon epithelium and colorectal carcinomas. We also investigated the association of mGluR4 expression levels with various clinicopathologic parameters. EXPERIMENTAL DESIGN mGluR4 expression was investigated in 21 normal and 312 malignant tissues from various organs using immunohistochemistry. In addition, 241 cases of colorectal carcinomas were examined and correlations between mGluR4 expression and various clinicopathologic parameters were then statistically analyzed. RESULTS Expression of mGluR4 was identified in the normal epithelia of the upper respiratory tract, gastrointestinal tracts, breast, uterine cervix, urinary bladder, and skin, whereas it was not detected in the thyroid, lung alveoli, liver, testis, or prostate. In the corresponding malignant tissues, mGluR4 expression was frequently identified in colorectal carcinoma (68%), followed by malignant melanoma, laryngeal carcinoma, and breast carcinomas. Expression of mGluR4 was detected in 131 (54%) of 241 colorectal carcinomas and 12 (5%) cases among them showed overexpression in their cytoplasms. Loss of mGluR4 expression was negatively associated with tumor differentiation (P = 0.028), whereas overexpression of mGluR4 was positively associated with recurrence (P = 0.034) and poor disease-free survival (P = 0.017) in multivariate analyses. CONCLUSIONS Our results suggest that mGluR4 signaling may play a role in colorectal carcinomas and that overexpression of mGluR4 is associated with poor prognosis.
Collapse
Affiliation(s)
- Hee Jin Chang
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | | | | | | | | | | |
Collapse
|
33
|
Matrisciano F, Scaccianoce S, Del Bianco P, Panaccione I, Canudas AM, Battaglia G, Riozzi B, Ngomba RT, Molinaro G, Tatarelli R, Melchiorri D, Nicoletti F. Metabotropic glutamate receptors and neuroadaptation to antidepressants: imipramine-induced down-regulation of beta-adrenergic receptors in mice treated with metabotropic glutamate 2/3 receptor ligands. J Neurochem 2005; 93:1345-52. [PMID: 15934953 DOI: 10.1111/j.1471-4159.2005.03141.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Antidepressant drugs have a clinical latency that correlates with the development of neuroadaptive changes, including down-regulation of beta-adrenergic receptors in different brain regions. The identification of drugs that shorten this latency will have a great impact on the treatment of major depressive disorders. We report that the time required for the antidepressant imipramine to reduce the expression of beta-adrenergic receptors in the hippocampus is reduced by a co-administration with centrally active ligands of type 2/3 metabotropic glutamate (mGlu2/3) receptors. Daily treatment of mice with imipramine alone (10 mg/kg, i.p.) reduced the expression of beta-adrenergic receptors in the hippocampus after 21 days, but not at shorter times, as assessed by western blot analysis of beta1-adrenergic receptors and by the amount of specifically bound [3H]CGP-12177, a selective beta-adrenergic receptor ligand. Down-regulation of beta-adrenergic receptors occurred at shorter times (i.e. after 14 days) when imipramine was combined with low doses (0.5 mg/kg, i.p.) of the selective mGlu2/3 receptor agonist LY379268, or with the preferential mGlu2/3 receptor antagonist LY341495 (1 mg/kg, i.p.). Higher doses of LY379268 (2 mg/kg, i.p.) were inactive. This intriguing finding suggests that neuroadaptation to imipramine--at least as assessed by changes in the expression of beta1-adrenergic receptors--is influenced by drugs that interact with mGlu2/3 receptors and stimulates further research aimed at establishing whether any of these drugs can shorten the clinical latency of classical antidepressants.
Collapse
MESH Headings
- Adaptation, Physiological
- Amino Acids/pharmacology
- Animals
- Antidepressive Agents, Tricyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Down-Regulation
- Excitatory Amino Acid Antagonists/pharmacology
- Hippocampus/metabolism
- Imipramine/pharmacology
- Ligands
- Male
- Mice
- Mice, Inbred Strains
- Nervous System Physiological Phenomena
- Reaction Time/drug effects
- Receptors, Adrenergic, beta/metabolism
- Receptors, Metabotropic Glutamate/administration & dosage
- Receptors, Metabotropic Glutamate/agonists
- Receptors, Metabotropic Glutamate/antagonists & inhibitors
- Receptors, Metabotropic Glutamate/metabolism
- Xanthenes/pharmacology
Collapse
Affiliation(s)
- F Matrisciano
- Department of Human Physiology and Pharmacology, University of Rome La Sapienza, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bhattacharya M, Babwah AV, Godin C, Anborgh PH, Dale LB, Poulter MO, Ferguson SSG. Ral and phospholipase D2-dependent pathway for constitutive metabotropic glutamate receptor endocytosis. J Neurosci 2005; 24:8752-61. [PMID: 15470141 PMCID: PMC6729950 DOI: 10.1523/jneurosci.3155-04.2004] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
G-protein-coupled receptors play a central role in the regulation of neuronal cell communication. Class 1 metabotropic glutamate receptors (mGluRs) mGluR1a and mGluR5a, which are coupled with the hydrolysis of phosphoinositides, are essential for modulating excitatory neurotransmission at glutamatergic synapses. These receptors are constitutively internalized in heterologous cell cultures, neuronal cultures, and intact neuronal tissues. We show here that the small GTP-binding protein Ral, its guanine nucleotide exchange factor RalGDS (Ral GDP dissociation stimulator), and phospholipase D2 (PLD2) are constitutively associated with class 1 mGluRs and regulate constitutive mGluR endocytosis. Moreover, both Ral and PLD2 are colocalized with mGluRs in endocytic vesicles in both human embryonic kidney 293 (HEK 293) cells and neurons. Ral and PLD2 activity is required for the internalization of class 1 mGluRs but is not required for the internalization of the beta2-adrenergic receptor. Constitutive class 1 mGluR internalization is not dependent on the downstream Ral effector proteins Ral-binding protein 1 and PLD1 or either ADP-ribosylation factors ARF1 or ARF6. The treatment of HEK 293 cells and neurons with small interfering RNA both downregulates PLD2 expression and blocks mGluR1a and mGluR5a endocytosis. The constitutive internalization of mGluR1a and mGluR5a is also attenuated by the treatment of cells with 1-butanol to prevent PLD2-mediated phosphatidic acid formation. We propose that the formation of a mGluR-scaffolded RalGDS/Ral/PLD2 protein complex provides a novel alternative mechanism to beta-arrestins for the constitutive endocytosis of class 1 mGluRs.
Collapse
Affiliation(s)
- Moshmi Bhattacharya
- Cell Biology Research Group, Robarts Research Institute, London, Ontario, Canada N6A 5K8
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The visual system can adjust its sensitivity over a wide range of light intensities. Photoreceptors account for some of this adjustment, but there is evidence that postreceptoral processes also exist. To investigate the latter, we pharmacologically mimicked the effects of light stimulation on mouse On bipolar cells, thus avoiding confounding effects of receptoral mechanisms. Here, we report that cGMP selectively enhances responses to dim, but not bright, stimuli through a purely postsynaptic mechanism. This action of cGMP was completely blocked by inhibitors of cGMP-dependent kinase. We propose that cGMP-dependent kinase decreases coupling of the On bipolar cell glutamate receptor to the downstream cascade, thus amplifying small decreases in photoreceptor transmitter levels that would otherwise go undetected by the visual system.
Collapse
Affiliation(s)
- Josefin Snellman
- Department of Neuroscience, Albert Einstein College of Medicine, The Rose F. Kennedy Center, Bronx, New York 10461, USA.
| | | |
Collapse
|
36
|
Lemon G, Brockhausen J, Li GH, Gibson WG, Bennett MR. Calcium mobilization and spontaneous transient outward current characteristics upon agonist activation of P2Y2 receptors in smooth muscle cells. Biophys J 2005; 88:1507-23. [PMID: 15556987 PMCID: PMC1305209 DOI: 10.1529/biophysj.104.043976] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2004] [Accepted: 08/12/2004] [Indexed: 11/18/2022] Open
Abstract
A quantitative model is provided that links the process of metabotropic receptor activation and sequestration to the generation of inositol 1,4,5-trisphosphate, the subsequent release of calcium from the central sarcoplasmic reticulum, and the consequent release of calcium from subsarcolemma sarcoplasmic reticulum that acts on large-conductance potassium channels to generate spontaneous transient outward currents (STOCs). This model is applied to the case of STOC generation in vascular A7r5 smooth muscle cells that have been transfected with a chimera of the P2Y(2) metabotropic receptor and green fluorescent protein (P2Y(2)-GFP) and exposed to the P2Y(2) receptor agonist uridine 5'-triphosphate. The extent of P2Y(2)-GFP sequestration from the membrane on exposure to uridine 5'-triphosphate, the ensuing changes in cytosolic calcium concentration, as well as the interval between STOCs that are subsequently generated, are used to determine parameter values in the model. With these values, the model gives a good quantitative prediction of the dynamic changes in STOC amplitude observed upon activation of metabotropic P2Y(2) receptors in the vascular smooth muscle cell line.
Collapse
MESH Headings
- Animals
- Biological Transport, Active/drug effects
- Biological Transport, Active/physiology
- Calcium/metabolism
- Calcium Signaling/drug effects
- Calcium Signaling/physiology
- Cell Line
- Computer Simulation
- Membrane Potentials/drug effects
- Membrane Potentials/physiology
- Models, Biological
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Purinergic P2 Receptor Agonists
- Receptors, Purinergic P2/metabolism
- Receptors, Purinergic P2Y2
- Uridine Triphosphate/pharmacology
Collapse
Affiliation(s)
- G Lemon
- The School of Mathematics and Statistics, University of Sydney, New South Wales, Australia
| | | | | | | | | |
Collapse
|
37
|
Pi M, Oakley RH, Gesty-Palmer D, Cruickshank RD, Spurney RF, Luttrell LM, Quarles LD. Beta-arrestin- and G protein receptor kinase-mediated calcium-sensing receptor desensitization. Mol Endocrinol 2005; 19:1078-87. [PMID: 15637145 DOI: 10.1210/me.2004-0450] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Extracellular calcium rapidly controls PTH secretion through binding to the G protein-coupled calcium-sensing receptor (CASR) expressed in parathyroid glands. Very little is known about the regulatory proteins involved in desensitization of CASR. G protein receptor kinases (GRK) and beta-arrestins are important regulators of agonist-dependent desensitization of G protein-coupled receptors. In the present study, we investigated their role in mediating agonist-dependent desensitization of CASR. In heterologous cell culture models, we found that the transfection of GRK4 inhibits CASR signaling by enhancing receptor phosphorylation and beta-arrestin translocation to the CASR. In contrast, we found that overexpression of GRK2 desensitizes CASR by classical mechanisms as well as through phosphorylation-independent mechanisms involving disruption of Galphaq signaling. In addition, we observed lower circulating PTH levels and an attenuated increase in serum PTH after hypocalcemic stimulation in beta-arrestin2 null mice, suggesting a functional role of beta-arrestin2-dependent desensitization pathways in regulating CASR function in vivo. We conclude that GRKs and beta-arrestins play key roles in regulating CASR responsiveness in parathyroid glands.
Collapse
Affiliation(s)
- Min Pi
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Bonsi P, Cuomo D, De Persis C, Centonze D, Bernardi G, Calabresi P, Pisani A. Modulatory action of metabotropic glutamate receptor (mGluR) 5 on mGluR1 function in striatal cholinergic interneurons. Neuropharmacology 2005; 49 Suppl 1:104-13. [PMID: 16005029 DOI: 10.1016/j.neuropharm.2005.05.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2005] [Revised: 05/12/2005] [Accepted: 05/12/2005] [Indexed: 11/21/2022]
Abstract
Within basal ganglia, group I metabotropic glutamate receptor subtypes (mGluR1 and 5) frequently co-localize in the same neuron. However, little is known about how these receptors functionally interact. We addressed this issue by means of electrophysiological recordings of striatal cholinergic interneurons, a neuronal subtype that co-express both group I mGluRs. The group I non-selective agonist 3,5-DHPG induced a membrane depolarization/inward current that was prevented by co-application of LY 367385, a selective mGluR1 antagonist, and SIB 1757 or MPEP, blockers of mGluR5 subtype. The reversal potential for the response to 3,5-DHPG was close to the equilibrium potential for potassium channels. Repeated bath or focal applications of 3,5-DHPG induced a progressive decline in the amplitude of the membrane depolarization, suggesting that group I mGluRs undergo receptor desensitization. Interestingly, in the presence of the mGluR5 blocker, SIB 1757, this event was not observed, whereas it occurred in LY 367385. PKC blockers chelerythrine and calphostin C mimicked the inhibitory effect of SIB 1757. In a subset of interneurons, in MPEP or SIB 1757, 3,5-DHPG induced a 0.5-1 Hz oscillatory response, that was prevented by L-type Ca2+ channel blockers, and by the tyrosine kinase inhibitors genistein and lavendustin. Together, these data suggest that mGluR5 modulates mGluR1 activity to shape cell excitability.
Collapse
Affiliation(s)
- P Bonsi
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, C.E.R.C., Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
39
|
Kushmerick C, Price GD, Taschenberger H, Puente N, Renden R, Wadiche JI, Duvoisin RM, Grandes P, von Gersdorff H. Retroinhibition of presynaptic Ca2+ currents by endocannabinoids released via postsynaptic mGluR activation at a calyx synapse. J Neurosci 2004; 24:5955-65. [PMID: 15229243 PMCID: PMC6729246 DOI: 10.1523/jneurosci.0768-04.2004] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We investigated the mechanisms by which activation of group I metabotropic glutamate receptors (mGluRs) and CB1 cannabinoid receptors (CB1Rs) leads to inhibition of synaptic currents at the calyx of Held synapse in the medial nucleus of the trapezoid body (MNTB) of the rat auditory brainstem. In approximately 50% of the MNTB neurons tested, activation of group I mGluRs by the specific agonist (s)-3,5-dihydroxyphenylglycine (DHPG) reversibly inhibited AMPA receptor- and NMDA receptor-mediated EPSCs to a similar extent and reduced paired-pulse depression, suggestive of an inhibition of glutamate release. Presynaptic voltage-clamp experiments revealed a reversible reduction of Ca2+ currents by DHPG, with no significant modification of the presynaptic action potential waveform. Likewise, in approximately 50% of the tested cells, the CB1 receptor agonist (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone (WIN) reversibly inhibited EPSCs, presynaptic Ca2+ currents, and exocytosis. For a given cell, the amount of inhibition by DHPG correlated with that by WIN. Moreover, the inhibitory action of DHPG was blocked by the CB1R antagonist N-(piperidin-1-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide (AM251) and occluded by WIN, indicating that DHPG and WIN operate via a common pathway. The inhibition of EPSCs by DHPG, but not by WIN, was abolished after dialyzing 40 mm BAPTA into the postsynaptic cell, suggesting that DHPG activated postsynaptic mGluRs. Light and electron microscopy immunolabeling indicated a presynaptic expression of CB1Rs and postsynaptic localization of mGluR1a. Our data suggest that activation of postsynaptic mGluRs triggers the Ca2+-dependent release of endocannabinoids that activate CB1 receptors on the calyx terminal, which leads to a reduction of presynaptic Ca2+ current and glutamate release.
Collapse
MESH Headings
- 2-Amino-5-phosphonovalerate/pharmacology
- Action Potentials/drug effects
- Amino Acids/pharmacology
- Animals
- Benzoxazines
- Brain Stem/drug effects
- Brain Stem/physiology
- Calcium Signaling/drug effects
- Cannabinoid Receptor Modulators/physiology
- Endocannabinoids
- Evoked Potentials, Auditory, Brain Stem/physiology
- Glycine/analogs & derivatives
- Glycine/pharmacology
- Ion Transport/drug effects
- Morpholines/pharmacology
- Naphthalenes/pharmacology
- Nerve Endings/drug effects
- Nerve Endings/physiology
- Patch-Clamp Techniques
- Picrotoxin/pharmacology
- Piperidines/pharmacology
- Pyrazoles/pharmacology
- Quinoxalines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/physiology
- Receptors, Metabotropic Glutamate/drug effects
- Receptors, Metabotropic Glutamate/physiology
- Receptors, Presynaptic/drug effects
- Receptors, Presynaptic/physiology
- Resorcinols/pharmacology
- Scopolamine/pharmacology
- Synaptic Transmission/drug effects
- Xanthenes/pharmacology
Collapse
|
40
|
Montiel M, Quesada J, Jiménez E. Activation of second messenger-dependent protein kinases induces muscarinic acetylcholine receptor desensitization in rat thyroid epithelial cells. Mol Cell Endocrinol 2004; 223:35-41. [PMID: 15279909 DOI: 10.1016/j.mce.2004.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2004] [Revised: 04/20/2004] [Accepted: 05/27/2004] [Indexed: 10/26/2022]
Abstract
Internalization and phosphorylation of G protein-coupled receptors (GPCR) are considered two important regulatory events of receptor signal transduction. In Fischer rat thyroid (FRT) epithelial cells, we have shown that muscarinic acetylcholine receptor (mAChR) stimulation induces intracellular Ca2+ mobilization via Ca2+ store release, capacitative Ca2+ entry and voltage-dependent Ca2+ channels activation. In the present study, the role of mAChR internalization and phosphorylation on receptor signalling pathway was examined by means of intracellular Ca2+ measurement in these cells. Exposure of FRT cells to carbachol (Cch), a mAChR agonist, resulted in a desensitization of receptor-mediated intracellular Ca2+ mobilization and induced the internalization of constitutively expressed mAChR in this cell type. Treatment of FRT cells with hypertonic sucrose, which markedly reduced agonist-receptor complex internalization, or phenylarsine oxide (PAO) diminished the Cch-induced intracellular Ca2+ response. Moreover, pretreatment of cells with phorbol-12-myristate-13-acetate (PMA), an activator of protein kinase C (PKC), completely abolished Cch-evoked Ca2+ mobilization, whereas it was significantly increased by the preincubation of cells with GF109203X, a selective inhibitor of PKC. We also found a marked decrease on Cch-stimulated Ca2+ mobilization in pretreated FRT cells with forskolin, an activator of protein kinase A (PKA), but the preincubation of cells with genistein, an inhibitor of protein tyrosine kinases, had no effect on Ca2+ mobilization induced by Cch. These findings seem to indicate that mAChR in FRT cells exhibit a desensitization, which may be mediated, at least in part, through activation of second messenger-dependent protein kinases and that receptor internalization could be necessary for signalling.
Collapse
Affiliation(s)
- Mercedes Montiel
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Málaga, Campus de Teatinos s/n, 29071 Málaga, Spain
| | | | | |
Collapse
|
41
|
Ismayilova N, Crossman A, Verkhratsky A, Brotchie J. Effects of adenosine A1, dopamine D1 and metabotropic glutamate 5 receptors-modulating agents on locomotion of the reserpinised rats. Eur J Pharmacol 2004; 497:187-95. [PMID: 15306204 DOI: 10.1016/j.ejphar.2004.06.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2004] [Accepted: 06/18/2004] [Indexed: 11/24/2022]
Abstract
The pathophysiology of Parkinson's disease and l-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesia are characterised by an imbalance between activity of the direct and indirect pathways regulated by dopamine D1 and D2 receptors, respectively. In this study, we investigated the effects of treatments combining adenosine A(1) and metabotropic glutamate 5 (mGlu5) receptors modulators on locomotion induced by dopamine D1 receptor activation in the reserpine-treated rats. Administration of the adenosine A(1) receptor agonist and mGlu5 receptor antagonist resulted in the significant reduction of dopamine D1 receptor agonist-induced locomotion. The combination of adenosine A(1) receptor agonist with mGlu5 receptor antagonist had no greater effect than these compounds alone. However, the adenosine A(1) receptor antagonist attenuated the inhibitory effect of mGlu5 receptor antagonist. The data suggest that the effect of mGlu5 receptor blockade on locomotion elicited by dopamine D1 receptor stimulation involves activation of adenosine A(1) receptors. This interaction can improve our understanding of pathophysiology of L-DOPA-induced dyskinesia.
Collapse
Affiliation(s)
- Naila Ismayilova
- The University of Manchester, School of Biological Sciences, 1.124 Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | | | | | | |
Collapse
|
42
|
Pacheco R, Ciruela F, Casadó V, Mallol J, Gallart T, Lluis C, Franco R. Group I metabotropic glutamate receptors mediate a dual role of glutamate in T cell activation. J Biol Chem 2004; 279:33352-8. [PMID: 15184389 DOI: 10.1074/jbc.m401761200] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Metabotropic glutamate receptors (mGluR) are present in cells of the nervous system, where they are activated by one of the main neurotransmitters, glutamate. They are also expressed in cells outside the nervous system. We identified and characterized two receptors belonging to group I mGluR, mGlu1R and mGlu5R, in human cell lines of lymphoid origin and in resting and activated lymphocytes from human peripheral blood. Both are highly expressed in the human Jurkat T cell line, whereas mGlu5R is expressed only in the human B cell line SKW6.4. In blood lymphocytes, mGlu5R is expressed constitutively, whereas mGlu1R is expressed only upon activation via the T cell receptor-CD3 complex. Group I receptors in the central nervous system are coupled to phospholipase C, whereas in blood lymphocytes, activation of mGlu5R does not trigger this signaling pathway, but instead activates adenylate cyclase. On the other hand, mGlu5R does not mediate ERK1/2 activation, whereas mGlu1R, which is coupled neither to phospholipase C nor to calcium channels and whose activation does not increase cAMP, activates the mitogen-activated protein kinase cascade. The differential expression of mGluR in resting and activated lymphocytes and the different signaling pathways that are triggered when mGlu1Rs or mGlu5Rs are activated point to a key role of glutamate in the regulation of T cell physiological function. The study of the signaling pathways (cAMP production and ERK1/2 phosphorylation) and the proliferative response obtained in the presence of glutamate analogs suggests that mGlu1R and mGlu5R have distinct functions. mGlu5R mediates the reported inhibition of cell proliferation evoked by glutamate, which is reverted by the activation of inducible mGlu1R. This is a novel non-inhibitory action mechanism for glutamate in lymphocyte activation. mGlu1R and mGlu5R thus mediate opposite glutamate effects in human lymphocytes.
Collapse
Affiliation(s)
- Rodrigo Pacheco
- Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
43
|
Pula G, Mundell SJ, Roberts PJ, Kelly E. Agonist-independent internalization of metabotropic glutamate receptor 1a is arrestin- and clathrin-dependent and is suppressed by receptor inverse agonists. J Neurochem 2004; 89:1009-20. [PMID: 15140199 DOI: 10.1111/j.1471-4159.2004.02387.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Three group I mGluR antagonists CPCCOEt, LY367385 and BAY36-7620, were analyzed for their effect on cell surface expression of metabotropic glutamate receptor 1a and 1b. All three antagonists inhibited glutamate-induced internalization of mGluR1a and mGluR1b. However, when added alone, either LY367385 or BAY36-7620 increased the cell surface expression of mGluR1a but not mGluR1b. Both LY367385 and BAY36-7620 displayed inverse agonist activity as judged by their ability to inhibit basal inositol phosphate accumulation in cells expressing the constitutively active mGluR1a. Interestingly, mGluR1a but not mGluR1b was constitutively internalized in HEK293 cells and both LY367385 and BAY36-7620 inhibited the constitutive internalization of this splice variant. Furthermore, coexpression of dominant negative mutant constructs of arrestin-2 [arrestin-2-(319-418)] or Eps15 [Eps15(E Delta 95-295)] increased cell surface expression of mGluR1a and blocked constitutive receptor internalization. In the presence of these dominant negative mutants, incubation of cells with LY367385 and BAY36-7620 produced no further increase in cell surface expression of mGluR1a. Taken together, these results suggest that the constitutive activity of mGluR1a triggers the internalization of the receptor through an arrestin- and clathrin-dependent pathway, and that inverse agonists increase the cell surface expression of mGluR1a by promoting an inactive form of mGluR1a, which does not undergo constitutive internalization.
Collapse
Affiliation(s)
- Giordano Pula
- Department of Pharmacology, School of Medical Sciences, University of Bristol, Bristol, UK
| | | | | | | |
Collapse
|
44
|
Yamaguchi K, Watanabe K. Pursuit of roles for metabotropic glutamate receptors in the anteroventral third ventricular region in regulating vasopressin secretion and cardiovascular function in conscious rats. Brain Res Bull 2004; 63:321-9. [PMID: 15196658 DOI: 10.1016/j.brainresbull.2004.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Revised: 03/31/2004] [Accepted: 04/01/2004] [Indexed: 11/22/2022]
Abstract
This study aimed to evaluate the roles of metabotropic glutamate receptors (mGluRs) in the anteroventral third ventricular region (AV3V; a pivotal area for osmotic responses and PGE2 actions) in regulating AVP secretion and cardiovascular function. In conscious and unrestrained rats, we examined the effects of AV3V infusion of t-ACPD (an agonist for mGluRs) and 8-bromo (Br)-cAMP (an agonist for cAMP associated with mGluR action) on plasma and cardiovascular variables, and the effects of MCPG (an antagonist for mGluRs) on the responses to t-ACPD, PGE2, and hyperosmolality. AV3V infusion of t-ACPD or 8-Br-cAMP produced dose-dependent rises in plasma AVP, arterial pressure and heart rate after 5 or 15 min, without altering plasma osmolality, sodium, potassium or chloride. t-ACPD administration into the cerebral ventricle had no effects on the variables. The plasma AVP and arterial pressure responses to AV3V t-ACPD infusion were blocked by preadministration of MCPG 15 min before the infusion. MCPG treatment was also potent at inhibiting the augmentation of plasma AVP elicited by AV3V infusion of PGE2, although its pressor and tachycardiac actions were not influenced. MCPG application, however, had no effect on either the increases in plasma AVP or arterial pressure in response to the enhanced plasma osmolality induced by i.v. infusion of hypertonic saline or their stable levels during isotonic saline infusion. Histological analysis showed that the AV3V drug infusion sites were located in structures such as the median or medial preoptic nucleus and periventricular nucleus. These results suggest that AV3V mGluRs may act to potentiate AVP release and cardiovascular function when stimulated in the basal state, and may participate in the hormone secretion prompted by AV3V PGE2, despite probable negligible contributions to the mechanisms responsible for the PGE2 cardiovascular effects or the phenomenon provoked by osmotic load.
Collapse
Affiliation(s)
- Ken'ichi Yamaguchi
- Department of Homeostatic Regulation and Development, Niigata University Graduate School of Medical and Dental Sciences, Asahimachi-Dori 1-757, Niigata City 951-8510, Japan.
| | | |
Collapse
|
45
|
Sterne-Marr R, Dhami GK, Tesmer JJG, Ferguson SSG. Characterization of GRK2 RH domain-dependent regulation of GPCR coupling to heterotrimeric G proteins. Methods Enzymol 2004; 390:310-36. [PMID: 15488186 DOI: 10.1016/s0076-6879(04)90020-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Heterotrimeric guanine nucleotide (G)-coupled receptors (GPCRs) form the largest family of integral membrane proteins. GPCR activation by an agonist promotes the exchange of GDP for GTP on the Galpha subunit of the heterotrimeric G protein. The dissociated Galpha and Gbetagamma subunits subsequently modulate the activity of a diverse assortment of effector systems. GPCR signaling via heterotrimeric G proteins is attenuated rapidly by the engagement of protein kinases. The canonical model for GPCR desensitization involves G protein-coupled receptor kinase (GRK)-dependent receptor phosphorylation to promote the binding of arrestin proteins that function to sterically block receptor:G-protein interactions. GRK2 and GRK3 have been shown to interact with Galphaq via the regulator of G-protein signaling (RGS) homology (RH) domain localized within their amino-terminal domains. It now appears that the G-protein uncoupling of many GPCRs linked to Galphaq, in particularly metabotropic glutamate receptors, may be mediated by the GRK2 RH domain via a phosphorylation-independent mechanism. This article reviews much of the background and methodology required for the characterization of the GRK2 phosphorylation-independent attenuation of GPCR signaling.
Collapse
|
46
|
Barbas D, DesGroseillers L, Castellucci VF, Carew TJ, Marinesco S. Multiple serotonergic mechanisms contributing to sensitization in aplysia: evidence of diverse serotonin receptor subtypes. Learn Mem 2003; 10:373-86. [PMID: 14557610 PMCID: PMC218003 DOI: 10.1101/lm.66103] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The neurotransmitter serotonin (5-HT) plays an important role in memory encoding in Aplysia. Early evidence showed that during sensitization, 5-HT activates a cyclic AMP-protein kinase A (cAMP-PKA)-dependent pathway within specific sensory neurons (SNs), which increases their excitability and facilitates synaptic transmission onto their follower motor neurons (MNs). However, recent data suggest that serotonergic modulation during sensitization is more complex and diverse. The neuronal circuits mediating defensive reflexes contain a number of interneurons that respond to 5-HT in ways opposite to those of the SNs, showing a decrease in excitability and/or synaptic depression. Moreover, in addition to acting through a cAMP-PKA pathway within SNs, 5-HT is also capable of activating a variety of other protein kinases such as protein kinase C, extracellular signal-regulated kinases, and tyrosine kinases. This diversity of 5-HT responses during sensitization suggests the presence of multiple 5-HT receptor subtypes within the Aplysia central nervous system. Four 5-HT receptors have been cloned and characterized to date. Although several others probably remain to be characterized in molecular terms, especially the Gs-coupled 5-HT receptor capable of activating cAMP-PKA pathways, the multiplicity of serotonergic mechanisms recruited into action during learning in Aplysia can now be addressed from a molecular point of view.
Collapse
Affiliation(s)
- Demian Barbas
- Département de biochimie, Université de Montréal, Québec H3C 3J7, Canada
| | | | | | | | | |
Collapse
|
47
|
Agnati LF, Ferré S, Lluis C, Franco R, Fuxe K. Molecular mechanisms and therapeutical implications of intramembrane receptor/receptor interactions among heptahelical receptors with examples from the striatopallidal GABA neurons. Pharmacol Rev 2003; 55:509-50. [PMID: 12869660 DOI: 10.1124/pr.55.3.2] [Citation(s) in RCA: 245] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The molecular basis for the known intramembrane receptor/receptor interactions among G protein-coupled receptors was postulated to be heteromerization based on receptor subtype-specific interactions between different types of receptor homomers. The discovery of GABAB heterodimers started this field rapidly followed by the discovery of heteromerization among isoreceptors of several G protein-coupled receptors such as delta/kappa opioid receptors. Heteromerization was also discovered among distinct types of G protein-coupled receptors with the initial demonstration of somatostatin SSTR5/dopamine D2 and adenosine A1/dopamine D1 heteromeric receptor complexes. The functional meaning of these heteromeric complexes is to achieve direct or indirect (via adapter proteins) intramembrane receptor/receptor interactions in the complex. G protein-coupled receptors also form heteromeric complexes involving direct interactions with ion channel receptors, the best example being the GABAA/dopamine D5 receptor heteromerization, as well as with receptor tyrosine kinases and with receptor activity modulating proteins. As an example, adenosine, dopamine, and glutamate metabotropic receptor/receptor interactions in the striatopallidal GABA neurons are discussed as well as their relevance for Parkinson's disease, schizophrenia, and drug dependence. The heterodimer is only one type of heteromeric complex, and the evidence is equally compatible with the existence of higher order heteromeric complexes, where also adapter proteins such as homer proteins and scaffolding proteins can exist. These complexes may assist in the process of linking G protein-coupled receptors and ion channel receptors together in a receptor mosaic that may have special integrative value and may constitute the molecular basis for some forms of learning and memory.
Collapse
Affiliation(s)
- Luigi F Agnati
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
48
|
Moldrich RX, Chapman AG, De Sarro G, Meldrum BS. Glutamate metabotropic receptors as targets for drug therapy in epilepsy. Eur J Pharmacol 2003; 476:3-16. [PMID: 12969743 DOI: 10.1016/s0014-2999(03)02149-6] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Metabotropic glutamate (mGlu) receptors have multiple actions on neuronal excitability through G-protein-linked modifications of enzymes and ion channels. They act presynaptically to modify glutamatergic and gamma-aminobutyric acid (GABA)-ergic transmission and can contribute to long-term changes in synaptic function. The recent identification of subtype-selective agonists and antagonists has permitted evaluation of mGlu receptors as potential targets in the treatment of epilepsy. Agonists acting on group I mGlu receptors (mGlu1 and mGlu5) are convulsant. Antagonists acting on mGlu1 or mGlu5 receptors are anticonvulsant against 3,5-dihydroxyphenylglycine (DHPG)-induced seizures and in mouse models of generalized motor seizures and absence seizures. The competitive, phenylglycine mGlu1/5 receptor antagonists generally require intracerebroventricular administration for potent anticonvulsant efficacy but noncompetitive antagonists, e.g., (3aS,6aS)-6a-naphthalen-2-ylmethyl-5-methyliden-hexahydrocyclopenta[c]furan-1-on (BAY36-7620), 2-methyl-6-(phenylethynyl)pyridine hydrochloride (MPEP), and 2-methyl-6-(2-phenylethenyl)pyridine (SIB-1893) block generalized seizures with systemic administration. Agonists acting on group II mGlu receptors (mGlu2, mGlu3) to reduce glutamate release are anticonvulsant, e.g., 2R,4R-aminopyrrolidine-2,4-dicarboxylate [(2R,4R)-APDC], (+)-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylic acid (LY354740), and (-)-2-oxa-4-aminobicyclo[3.1.0]hexane-4,6-dicarboxylate (LY379268). The classical agonists acting on group III mGlu receptors such as L-(+)-2-amino-4-phosphonobutyric acid, and L-serine-O-phosphate are acutely proconvulsant with some anticonvulsant activity. The more recently identified agonists (R,S)-4-phosphonophenylglycine [(R,S)-PPG] and (S)-3,4-dicarboxyphenylglycine [(S)-3,4-DCPG] and (1S,3R,4S)-1-aminocyclopentane-1,2,4-tricarboxylic acid [ACPT-1] are all anticonvulsant without proconvulsant effects. Studies in animal models of kindling reveal some efficacy of mGlu receptor ligands against fully kindled limbic seizures. In genetic mouse models, mGlu1/5 antagonists and mGlu2/3 agonists are effective against absence seizures. Thus, antagonists at group I mGlu receptors and agonists at groups II and III mGlu receptors are potential antiepileptic agents, but their clinical usefulness will depend on their acute and chronic side effects. Potential also exists for combining mGlu receptor ligands with other glutamatergic and non-glutamatergic agents to produce an enhanced anticonvulsant effect. This review also discusses what is known about mGlu receptor expression and function in rodent epilepsy models and human epileptic conditions.
Collapse
Affiliation(s)
- Randal X Moldrich
- Department of Pharmacology, Monash University, Melbourne 3800, Australia
| | | | | | | |
Collapse
|