1
|
Gawel K. A Review on the Role and Function of Cinnabarinic Acid, a "Forgotten" Metabolite of the Kynurenine Pathway. Cells 2024; 13:453. [PMID: 38474418 DOI: 10.3390/cells13050453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
In the human body, the majority of tryptophan is metabolized through the kynurenine pathway. This consists of several metabolites collectively called the kynurenines and includes, among others, kynurenic acid, L-kynurenine, or quinolinic acid. The wealth of metabolites, as well as the associated molecular targets and biological pathways, bring about a situation wherein even a slight imbalance in the kynurenine levels, both in the periphery and central nervous system, have broad consequences regarding general health. Cinnabarinic acid (CA) is the least known trace kynurenine, and its physiological and pathological roles are not widely understood. Some studies, however, indicate that it might be neuroprotective. Information on its hepatoprotective properties have also emerged, although these are pioneering studies and need to be replicated. Therefore, in this review, I aim to present and critically discuss the current knowledge on CA and its role in physiological and pathological settings to guide future studies.
Collapse
Affiliation(s)
- Kinga Gawel
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego 8b Str., 20-090 Lublin, Poland
| |
Collapse
|
2
|
Zhu C, Lan X, Wei Z, Yu J, Zhang J. Allosteric modulation of G protein-coupled receptors as a novel therapeutic strategy in neuropathic pain. Acta Pharm Sin B 2024; 14:67-86. [PMID: 38239234 PMCID: PMC10792987 DOI: 10.1016/j.apsb.2023.07.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/09/2023] [Accepted: 07/12/2023] [Indexed: 01/22/2024] Open
Abstract
Neuropathic pain is a debilitating pathological condition that presents significant therapeutic challenges in clinical practice. Unfortunately, current pharmacological treatments for neuropathic pain lack clinical efficacy and often lead to harmful adverse reactions. As G protein-coupled receptors (GPCRs) are widely distributed throughout the body, including the pain transmission pathway and descending inhibition pathway, the development of novel neuropathic pain treatments based on GPCRs allosteric modulation theory is gaining momentum. Extensive research has shown that allosteric modulators targeting GPCRs on the pain pathway can effectively alleviate symptoms of neuropathic pain while reducing or eliminating adverse effects. This review aims to provide a comprehensive summary of the progress made in GPCRs allosteric modulators in the treatment of neuropathic pain, and discuss the potential benefits and adverse factors of this treatment. We will also concentrate on the development of biased agonists of GPCRs, and based on important examples of biased agonist development in recent years, we will describe universal strategies for designing structure-based biased agonists. It is foreseeable that, with the continuous improvement of GPCRs allosteric modulation and biased agonist theory, effective GPCRs allosteric drugs will eventually be available for the treatment of neuropathic pain with acceptable safety.
Collapse
Affiliation(s)
- Chunhao Zhu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Xiaobing Lan
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Zhiqiang Wei
- Medicinal Chemistry and Bioinformatics Center, Ocean University of China, Qingdao 266100, China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jian Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
3
|
Xie RG, Xu GY, Wu SX, Luo C. Presynaptic glutamate receptors in nociception. Pharmacol Ther 2023; 251:108539. [PMID: 37783347 DOI: 10.1016/j.pharmthera.2023.108539] [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: 05/26/2023] [Revised: 08/19/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Chronic pain is a frequent, distressing and poorly understood health problem. Plasticity of synaptic transmission in the nociceptive pathways after inflammation or injury is assumed to be an important cellular basis for chronic, pathological pain. Glutamate serves as the main excitatory neurotransmitter at key synapses in the somatosensory nociceptive pathways, in which it acts on both ionotropic and metabotropic glutamate receptors. Although conventionally postsynaptic, compelling anatomical and physiological evidence demonstrates the presence of presynaptic glutamate receptors in the nociceptive pathways. Presynaptic glutamate receptors play crucial roles in nociceptive synaptic transmission and plasticity. They modulate presynaptic neurotransmitter release and synaptic plasticity, which in turn regulates pain sensitization. In this review, we summarize the latest understanding of the expression of presynaptic glutamate receptors in the nociceptive pathways, and how they contribute to nociceptive information processing and pain hypersensitivity associated with inflammation / injury. We uncover the cellular and molecular mechanisms of presynaptic glutamate receptors in shaping synaptic transmission and plasticity to mediate pain chronicity, which may provide therapeutic approaches for treatment of chronic pain.
Collapse
Affiliation(s)
- Rou-Gang Xie
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Sheng-Xi Wu
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Ceng Luo
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
4
|
Li X, Liu D, Dai Z, You Y, Chen Y, Lei C, Lv Y, Wang Y. Intraperitoneal 5-Azacytidine Alleviates Nerve Injury-Induced Pain in Rats by Modulating DNA Methylation. Mol Neurobiol 2023; 60:2186-2199. [PMID: 36627549 DOI: 10.1007/s12035-022-03196-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/23/2022] [Indexed: 01/12/2023]
Abstract
To investigate the role of DNA methylation in modulating chronic neuropathic pain (NPP), identify possible target genes of DNA methylation involved in this process, and preliminarily confirm the medicinal value of the DNA methyltransferases (DNMTs) inhibitor 5-azacytidine (5-AZA) in NPP by targeting gene methylation. Two rat NPP models, chronic constriction injury (CCI) and spinal nerve ligation (SNL), were used. The DNA methylation profiles in the lumbar spinal cord were assayed using an Arraystar Rat RefSeq Promoter Array. The underlying genes with differential methylation were then identified and submitted to Gene Ontology and pathway analysis. Methyl-DNA immunoprecipitation quantitative PCR (MeDIP-qPCR) and quantitative reverse transcription-PCR (RT-qPCR) were used to confirm gene methylation and expression. The protective function of 5-AZA in NPP and gene expression were evaluated via behavioral assays and RT-qPCR, respectively. Analysis of the DNA methylation patterns in the lumbar spinal cord indicated that 1205 differentially methylated fragments in CCI rats were located within DNA promoter regions, including 638 hypermethylated fragments and 567 hypomethylated fragments. The methylation levels of Grm4, Htr4, Adrb2, Kcnf1, Gad2, and Pparg, which are associated with long-term potentiation (LTP) and glutamatergic synapse pathways, were increased with a corresponding decrease in their mRNA expression, in the spinal cords of CCI rats. Moreover, we found that the intraperitoneal injection of 5-AZA (4 mg/kg) attenuated CCI- or SNL-induced mechanical allodynia and thermal hyperalgesia. Finally, the mRNA expression of hypermethylated genes such as Grm4, Htr4, Adrb2, Kcnf1, and Gad2 was reversed after 5-AZA treatment. CCI induced widespread methylation changes in the DNA promoter regions in the lumbar spinal cord. Intraperitoneal 5-AZA alleviated hyperalgesia in CCI and SNL rats, an effect accompanied by the reversed expression of hypermethylated genes. Thus, DNA methylation inhibition represents a promising epigenetic strategy for protection against chronic NPP following nerve injury. Our study lays a theoretical foundation for 5-AZA to become a clinical targeted drug.
Collapse
Affiliation(s)
- Xuan Li
- Department of Anesthesiology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - DeZhao Liu
- Department of Anesthesiology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - ZhiSen Dai
- Department of Anesthesiology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - YiSheng You
- Department of Anesthesiology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yan Chen
- Department of Anesthesiology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - ChenXing Lei
- Department of Anesthesiology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - YouYou Lv
- Department of Anesthesiology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Ying Wang
- Department of Anesthesiology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China. .,Department of Anesthesiology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
5
|
Pereira V, Arias JA, Llebaria A, Goudet C. Photopharmacological manipulation of amygdala metabotropic glutamate receptor mGlu4 alleviates neuropathic pain. Pharmacol Res 2023; 187:106602. [PMID: 36529205 DOI: 10.1016/j.phrs.2022.106602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
Neuropathic pain is a common health problem resulting in exacerbated response to noxious and non noxious stimuli, as well as impaired emotional and cognitive responses. Unfortunately, neuropathic pain is also one of the most difficult pain syndromes to manage, highlighting the importance of better understanding the brain regions and neuromodulatory mechanisms involved in its regulation. Among the many interconnected brain areas which process pain, the amygdala is known to play an important role in the integration of sensory and emotional pain signals. Here we questioned the ability of a recently identified neuromodulatory mechanism associated with the metabotropic glutamate receptors mGlu4 in the amygdala to modulate neuropathic pain. In a murine model of peripheral mononeuropathy, we demonstrate that pharmacological activation of amygdala mGlu4 efficiently alleviates sensory and depressive-like symptoms in both male and female mice. Moreover, we reveal a differential modulation of these symptoms. Activating mGlu4 in the contralateral amygdala relative to the side of the mononeuropathy, is necessary and sufficient to relieve both sensory and depressive-like symptoms, while ipsilateral activation solely reduces depressive-like symptoms. Furthermore, using photopharmacology, a recent strategy allowing precise photocontrol of endogenous proteins, we further demonstrate the dynamic alleviation of neuropathic pain through light-dependent facilitation of mGlu4 by a photoswitchable positive allosteric modulator. Finally, coupling photopharmacology and analgesic conditioned place preference, we show a significant pain-reducing effect of mGlu4 activation. Taken together, these data highlight the analgesic potential of enhancing amygdala mGlu4 activity to counteract neuropathy reinforcing its therapeutic interest for the treatment of pathological pain.
Collapse
Affiliation(s)
| | | | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Cyril Goudet
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
6
|
Mazzitelli M, Presto P, Antenucci N, Meltan S, Neugebauer V. Recent Advances in the Modulation of Pain by the Metabotropic Glutamate Receptors. Cells 2022; 11:2608. [PMID: 36010684 PMCID: PMC9406805 DOI: 10.3390/cells11162608] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 01/22/2023] Open
Abstract
Metabotropic glutamate receptors (mGluR or mGlu) are G-protein coupled receptors activated by the binding of glutamate, the main classical neurotransmitter of the nervous system. Eight different mGluR subtypes (mGluR1-8) have been cloned and are classified in three groups based on their molecular, pharmacological and signaling properties. mGluRs mediate several physiological functions such as neuronal excitability and synaptic plasticity, but they have also been implicated in numerous pathological conditions including pain. The availability of new and more selective allosteric modulators together with the canonical orthosteric ligands and transgenic technologies has led to significant advances in our knowledge about the role of the specific mGluR subtypes in the pathophysiological mechanisms of various diseases. Although development of successful compounds acting on mGluRs for clinical use has been scarce, the subtype-specific-pharmacological manipulation might be a compelling approach for the treatment of several disorders in humans, including pain; this review aims to summarize and update on preclinical evidence for the roles of different mGluRs in the pain system and discusses knowledge gaps regarding mGluR-related sex differences and neuroimmune signaling in pain.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Nico Antenucci
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Shakira Meltan
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
7
|
Notartomaso S, Boccella S, Antenucci N, Ricciardi F, Fazio F, Liberatore F, Scarselli P, Scioli M, Mascio G, Bruno V, Battaglia G, Nicoletti F, Maione S, Luongo L. Analgesic Activity of Cinnabarinic Acid in Models of Inflammatory and Neuropathic Pain. Front Mol Neurosci 2022; 15:892870. [PMID: 35721314 PMCID: PMC9204652 DOI: 10.3389/fnmol.2022.892870] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/05/2022] [Indexed: 12/02/2022] Open
Abstract
Cinnabarinic acid (CA) is a trace kynurenine metabolite, which activates both type-4 metabotropic glutamate (mGlu4) and arylic hydrocarbon (Ah) receptors. We examined the action of CA in models of inflammatory and neuropathic pain moving from the evidence that mGlu4 receptors are involved in the regulation of pain thresholds. Systemic administration of low doses of CA (0.125 and 0.25 mg/kg, i.p.) reduced nocifensive behaviour in the second phase of the formalin test. CA-induced analgesia was abrogated in mGlu4 receptor knockout mice, but was unaffected by treatment with the Ah receptor antagonist, CH223191 (1 mg/Kg, s.c.). Acute injection of low doses of CA (0.25 mg/kg, i.p.) also caused analgesia in mice subjected to Chronic Constriction Injury (CCI) of the sciatic nerve. Electrophysiological recording showed no effect of CA on spinal cord nociceptive neurons and a trend to a lowering effect on the frequency and duration of excitation of the rostral ventromedial medulla (RVM) ON cells in CCI mice. However, local application of CH223191 or the group-III mGlu receptor antagonist, MSOP disclosed a substantial lowering and enhancing effect of CA on both populations of neurons, respectively. When repeatedly administered to CCI mice, CA retained the analgesic activity only when combined with CH223191. Repeated administration of CA plus CH223191 restrained the activity of both spinal nociceptive neurons and RVM ON cells, in full agreement with the analgesic activity. These findings suggest that CA is involved in the regulation of pain transmission, and its overall effect depends on the recruitment of mGlu4 and Ah receptors.
Collapse
Affiliation(s)
- Serena Notartomaso
- Department of Molecular Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Serena Boccella
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - N. Antenucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Flavia Ricciardi
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesco Fazio
- Department of Psychiatry and Health Science, University of California, San Diego, La Jolla, CA, United States
| | - F. Liberatore
- Department of Molecular Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - P. Scarselli
- Department of Molecular Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - M. Scioli
- Department of Molecular Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Giada Mascio
- Department of Molecular Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - V. Bruno
- Department of Molecular Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Battaglia
- Department of Molecular Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Ferdinando Nicoletti
- Department of Molecular Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
- *Correspondence: Livio Luongo,
| |
Collapse
|
8
|
Palazzo E, Boccella S, Marabese I, Perrone M, Belardo C, Iannotta M, Scuteri D, De Dominicis E, Pagano M, Infantino R, Bagetta G, Maione S. Homo-AMPA in the periaqueductal grey modulates pain and rostral ventromedial medulla activity in diabetic neuropathic mice. Neuropharmacology 2022; 212:109047. [DOI: 10.1016/j.neuropharm.2022.109047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/22/2022] [Accepted: 03/26/2022] [Indexed: 12/21/2022]
|
9
|
Hofmann CS, Carrington S, Keller AN, Gregory KJ, Niswender CM. Regulation and functional consequences of mGlu 4 RNA editing. RNA (NEW YORK, N.Y.) 2021; 27:1220-1240. [PMID: 34244459 PMCID: PMC8457003 DOI: 10.1261/rna.078729.121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/01/2021] [Indexed: 06/13/2023]
Abstract
Metabotropic glutamate receptor 4 (mGlu4) is one of eight mGlu receptors within the Class C G protein-coupled receptor superfamily. mGlu4 is primarily localized to the presynaptic membrane of neurons where it functions as an auto and heteroreceptor controlling synaptic release of neurotransmitter. mGlu4 is implicated in numerous disorders and is a promising drug target; however, more remains to be understood about its regulation and pharmacology. Using high-throughput sequencing, we have validated and quantified an adenosine-to-inosine (A-to-I) RNA editing event that converts glutamine 124 to arginine in mGlu4; additionally, we have identified a rare but novel K129R site. Using an in vitro editing assay, we then validated the pre-mRNA duplex that allows for editing by ADAR enzymes and predicted its conservation across the mammalian species. Structural modeling of the mGlu4 protein predicts the Q124R substitution to occur in the B helix of the receptor that is critical for receptor dimerization and activation. Interestingly, editing of a receptor homodimer does not disrupt G protein activation in response to the endogenous agonist, glutamate. Using an assay designed to specifically measure heterodimer populations at the surface, however, we found that Q124R substitution decreased the propensity of mGlu4 to heterodimerize with mGlu2 and mGlu7 Our study is the first to extensively describe the extent and regulatory factors of RNA editing of mGlu4 mRNA transcripts. In addition, we have proposed a novel functional consequence of this editing event that provides insights regarding its effects in vivo and expands the regulatory capacity for mGlu receptors.
Collapse
MESH Headings
- Adenosine Deaminase/genetics
- Adenosine Deaminase/metabolism
- Amino Acid Sequence
- Animals
- Base Pairing
- Base Sequence
- Birds
- Cerebral Cortex/cytology
- Cerebral Cortex/metabolism
- Corpus Striatum/cytology
- Corpus Striatum/metabolism
- HEK293 Cells
- Hippocampus/cytology
- Hippocampus/metabolism
- Humans
- Models, Molecular
- Neurons/cytology
- Neurons/metabolism
- Nucleic Acid Conformation
- Point Mutation
- Protein Conformation, alpha-Helical
- Protein Conformation, beta-Strand
- RNA Editing
- RNA, Messenger/chemistry
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Metabotropic Glutamate/chemistry
- Receptors, Metabotropic Glutamate/genetics
- Receptors, Metabotropic Glutamate/metabolism
- Reptiles
- Sequence Homology, Amino Acid
Collapse
Affiliation(s)
- Christopher S Hofmann
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Sheridan Carrington
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Andrew N Keller
- Department of Pharmacology and Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Karen J Gregory
- Department of Pharmacology and Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| |
Collapse
|
10
|
A nanobody activating metabotropic glutamate receptor 4 discriminates between homo- and heterodimers. Proc Natl Acad Sci U S A 2021; 118:2105848118. [PMID: 34385321 DOI: 10.1073/pnas.2105848118] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There is growing interest in developing biologics due to their high target selectivity. The G protein-coupled homo- and heterodimeric metabotropic glutamate (mGlu) receptors regulate many synapses and are promising targets for the treatment of numerous brain diseases. Although subtype-selective allosteric small molecules have been reported, their effects on the recently discovered heterodimeric receptors are often not known. Here, we describe a nanobody that specifically and fully activates homodimeric human mGlu4 receptors. Molecular modeling and mutagenesis studies revealed that the nanobody acts by stabilizing the closed active state of the glutamate binding domain by interacting with both lobes. In contrast, this nanobody does not activate the heterodimeric mGlu2-4 but acts as a pure positive allosteric modulator. These data further reveal how an antibody can fully activate a class C receptor and bring further evidence that nanobodies represent an alternative way to specifically control mGlu receptor subtypes.
Collapse
|
11
|
Facilitating mGluR4 activity reverses the long-term deleterious consequences of chronic morphine exposure in male mice. Neuropsychopharmacology 2021; 46:1373-1385. [PMID: 33349673 PMCID: PMC8136479 DOI: 10.1038/s41386-020-00927-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022]
Abstract
Understanding the neurobiological underpinnings of abstinence from drugs of abuse is critical to allow better recovery and ensure relapse prevention in addicted subjects. By comparing the long-term transcriptional consequences of morphine and cocaine exposure, we identified the metabotropic glutamate receptor subtype 4 (mGluR4) as a promising pharmacological target in morphine abstinence. We evaluated the behavioral and molecular effects of facilitating mGluR4 activity in abstinent mice. Transcriptional regulation of marker genes of medium spiny neurons (MSNs) allowed best discriminating between 4-week morphine and cocaine abstinence in the nucleus accumbens (NAc). Among these markers, Grm4, encoding mGluR4, displayed down-regulated expression in the caudate putamen and NAc of morphine, but not cocaine, abstinent mice. Chronic administration of the mGluR4 positive allosteric modulator (PAM) VU0155041 (2.5 and 5 mg/kg) rescued social behavior, normalized stereotypies and anxiety and blunted locomotor sensitization in morphine abstinent mice. This treatment improved social preference but increased stereotypies in cocaine abstinent mice. Finally, the beneficial behavioral effects of VU0155041 treatment in morphine abstinent mice were correlated with restored expression of key MSN and neural activity marker genes in the NAc. This study reports that chronic administration of the mGluR4 PAM VU0155041 relieves long-term deleterious consequences of morphine exposure. It illustrates the neurobiological differences between opiate and psychostimulant abstinence and points to pharmacological repression of excessive activity of D2-MSNs in the NAc as a promising therapeutic lever in drug addiction.
Collapse
|
12
|
Wang X, Ali N, Lin CLG. Emerging role of glutamate in the pathophysiology and therapeutics of Gulf War illness. Life Sci 2021; 280:119609. [PMID: 33991547 DOI: 10.1016/j.lfs.2021.119609] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 11/20/2022]
Abstract
Gulf War illness (GWI) is a chronic and multi-symptomatic disorder affecting veterans who served in the Gulf War. The commonly reported symptoms in GWI veterans include mood problems, cognitive impairment, muscle and joint pain, migraine/headache, chronic fatigue, gastrointestinal complaints, skin rashes, and respiratory problems. Neuroimaging studies have revealed significant brain structure alterations in GWI veterans, including subcortical atrophy, decreased volume of the hippocampus, reduced total grey and white matter, and increased brain white matter axial diffusivity. These brain changes may contribute to or increase the severities of the GWI-related symptoms. Epidemiological studies have revealed that neurotoxic exposures and stress may be significant contributors to the development of GWI. However, the mechanism underlying how the exposure and stress could contribute to the multi-symptomatic disorder of GWI remains unclear. We and others have demonstrated that rodent models exposed to GW-related agents and stress exhibited higher extracellular glutamate levels, as well as impaired structure and function of glutamatergic synapses. Restoration of the glutamatergic synapses ameliorated the GWI-related pathological and behavioral deficits. Moreover, recent studies showed that a low-glutamate diet reduced multiple symptoms in GWI veterans, suggesting an important role of the glutamatergic system in GWI. Currently, growing evidence has indicated that abnormal glutamate neurotransmission may contribute to the GWI symptoms. This review summarizes the potential roles of glutamate dyshomeostasis and dysfunction of the glutamatergic system in linking the initial cause to the multi-symptomatic outcomes in GWI and suggests the glutamatergic system as a therapeutic target for GWI.
Collapse
Affiliation(s)
- Xueqin Wang
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Noor Ali
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Chien-Liang Glenn Lin
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
13
|
Gregory KJ, Goudet C. International Union of Basic and Clinical Pharmacology. CXI. Pharmacology, Signaling, and Physiology of Metabotropic Glutamate Receptors. Pharmacol Rev 2020; 73:521-569. [PMID: 33361406 DOI: 10.1124/pr.119.019133] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors respond to glutamate, the major excitatory neurotransmitter in the mammalian brain, mediating a modulatory role that is critical for higher-order brain functions such as learning and memory. Since the first mGlu receptor was cloned in 1992, eight subtypes have been identified along with many isoforms and splice variants. The mGlu receptors are transmembrane-spanning proteins belonging to the class C G protein-coupled receptor family and represent attractive targets for a multitude of central nervous system disorders. Concerted drug discovery efforts over the past three decades have yielded a wealth of pharmacological tools including subtype-selective agents that competitively block or mimic the actions of glutamate or act allosterically via distinct sites to enhance or inhibit receptor activity. Herein, we review the physiologic and pathophysiological roles for individual mGlu receptor subtypes including the pleiotropic nature of intracellular signal transduction arising from each. We provide a comprehensive analysis of the in vitro and in vivo pharmacological properties of prototypical and commercially available orthosteric agonists and antagonists as well as allosteric modulators, including ligands that have entered clinical trials. Finally, we highlight emerging areas of research that hold promise to facilitate rational design of highly selective mGlu receptor-targeting therapeutics in the future. SIGNIFICANCE STATEMENT: The metabotropic glutamate receptors are attractive therapeutic targets for a range of psychiatric and neurological disorders. Over the past three decades, intense discovery efforts have yielded diverse pharmacological tools acting either competitively or allosterically, which have enabled dissection of fundamental biological process modulated by metabotropic glutamate receptors and established proof of concept for many therapeutic indications. We review metabotropic glutamate receptor molecular pharmacology and highlight emerging areas that are offering new avenues to selectively modulate neurotransmission.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| | - Cyril Goudet
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| |
Collapse
|
14
|
Boccella S, Marabese I, Guida F, Luongo L, Maione S, Palazzo E. The Modulation of Pain by Metabotropic Glutamate Receptors 7 and 8 in the Dorsal Striatum. Curr Neuropharmacol 2020; 18:34-50. [PMID: 31210112 PMCID: PMC7327935 DOI: 10.2174/1570159x17666190618121859] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/01/2019] [Accepted: 05/31/2019] [Indexed: 12/28/2022] Open
Abstract
The dorsal striatum, apart from controlling voluntary movement, displays a recently demonstrated pain inhibition. It is connected to the descending pain modulatory system and in particular to the rostral ventromedial medulla through the medullary dorsal reticular nucleus. Diseases of the basal ganglia, such as Parkinson's disease, in addition to being characterized by motor disorders, are associated with pain and hyperactivation of the excitatory transmission. A way to counteract glutamatergic hyperactivation is through the activation of group III metabotropic glutamate receptors (mGluRs), which are located on presynaptic terminals inhibiting neurotransmitter release. So far the mGluRs of group III have been the least investigated, owing to a lack of selective tools. More recently, selective ligands for each mGluR of group III, in particular positive and negative allosteric modulators, have been developed and the role of each subtype is starting to emerge. The neuroprotective potential of group III mGluRs in pathological conditions, such as those characterized by elevate glutamate, has been recently shown. In the dorsal striatum, mGluR7 and mGluR8 are located at glutamatergic corticostriatal terminals and their stimulation inhibits pain in pathological conditions such as neuropathic pain. The two receptors in the dorsal striatum have instead a different role in pain control in normal conditions. This review will discuss recent results focusing on the contribution of mGluR7 and mGluR8 in the dorsal striatal control of pain. The role of mGluR4, whose antiparkinsonian activity is widely reported, will also be addressed.
Collapse
Affiliation(s)
- Serena Boccella
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Enza Palazzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
15
|
Okubo M, Yamanaka H, Kobayashi K, Noguchi K. Differential expression of mGluRs in rat spinal dorsal horns and their modulatory effects on nocifensive behaviors. Mol Pain 2019; 15:1744806919875026. [PMID: 31432760 PMCID: PMC6751533 DOI: 10.1177/1744806919875026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Glutamate is a neurotransmitter present in most excitatory synapses in the nervous system. It also plays a key role in the spinal cord’s physiological excitatory circuit and is involved in pathological neurotransmissions such as those observed in inflammatory and neuropathic pain conditions. The actions of glutamate are mediated by different types of ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs). Although expressions of iGluRs are well studied, those of mGluRs are not fully elucidated in the spinal cord. In this study, we examined the expressions of mGluRs (mGluR1-8) and investigated which mGluR subtypes can modulate pain transmission in the dorsal horn of the spinal cord using an inflammatory pain model. Reverse transcription-polymerase chain reaction revealed that mGluR mRNAs, except for mGluR2 and 6, were detected in the spinal cord. Double labeling analysis, in situ hybridization histochemistry with immunohistochemistry, was used to examine the distribution of each mGluR in neurons or glial cells in the lamina I–II of the spinal dorsal horn. mGluR1, 5, and 7 were generally, and 4 and 8 were frequently, expressed in neurons. mGluR3 was expressed not only in neurons but also in oligodendrocytes. We next examined the distribution of mGluR4 and 8 were expressed in excitatory or inhibitory neurons. Both mGluR4 and 8 were preferentially expressed in inhibitory neurons rather than in excitatory neurons. Furthermore, intrathecal delivery of CPPG((RS)-α-cyclopropyl-4-phosphonophenylglycine), an antagonist for mGluR 4 and 8, attenuated nocifensive behaviors and the increase in fos-positive-excitatory neurons of the dorsal horn induced by intraplantar injection of formalin. These findings suggest that mGluR4 and 8, which are preferentially expressed in inhibitory neurons, may play roles in the modulation of pain transmission in the spinal dorsal horn.
Collapse
Affiliation(s)
- Masamichi Okubo
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Hyogo, Japan
| | - Hiroki Yamanaka
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Hyogo, Japan
| | - Kimiko Kobayashi
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Hyogo, Japan
| | - Koichi Noguchi
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Hyogo, Japan
| |
Collapse
|
16
|
Abstract
In this issue of Cell Chemical Biology, the elegant manuscript by Rovira et al. (2016) describes a negative allosteric modulator (NAM) of mGlu4 metabotropic glutamate receptors with in vivo activity. This compound is rapidly and reversibly inactivated by light and represents a powerful pharmacological tool for the study of mGlu4 receptors in their native environment.
Collapse
Affiliation(s)
- Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University Sapienza of Rome, 00185 Rome, Italy; IRCCS Neuromed, 86077 Pozzilli, Italy.
| |
Collapse
|
17
|
Girard B, Tuduri P, Moreno MP, Sakkaki S, Barboux C, Bouschet T, Varrault A, Vitre J, McCort-Tranchepain I, Dairou J, Acher F, Fagni L, Marchi N, Perroy J, Bertaso F. The mGlu7 receptor provides protective effects against epileptogenesis and epileptic seizures. Neurobiol Dis 2019; 129:13-28. [PMID: 31051234 DOI: 10.1016/j.nbd.2019.04.016] [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: 01/10/2019] [Revised: 03/28/2019] [Accepted: 04/29/2019] [Indexed: 01/26/2023] Open
Abstract
Finding new targets to control or reduce seizure activity is essential to improve the management of epileptic patients. We hypothesized that activation of the pre-synaptic and inhibitory metabotropic glutamate receptor type 7 (mGlu7) reduces spontaneous seizures. We tested LSP2-9166, a recently developed mGlu7/4 agonist with unprecedented potency on mGlu7 receptors, in two paradigms of epileptogenesis. In a model of chemically induced epileptogenesis (pentylenetetrazole systemic injection), LSP2-9166 induces an anti-epileptogenic effect rarely observed in preclinical studies. In particular, we found a bidirectional modulation of seizure progression by mGlu4 and mGlu7 receptors, the latter preventing kindling. In the intra-hippocampal injection of kainic acid mouse model that mimics the human mesial temporal lobe epilepsy, we found that LSP2-9166 reduces seizure frequency and hippocampal sclerosis. LSP2-9166 also acts as an anti-seizure drug on established seizures in both models tested. Specific modulation of the mGlu7 receptor could represent a novel approach to reduce pathological network remodeling.
Collapse
Affiliation(s)
- Benoit Girard
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | - Pola Tuduri
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | | | - Sophie Sakkaki
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | | | | | - Annie Varrault
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | - Jihane Vitre
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | | | | | | | - Laurent Fagni
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | - Nicola Marchi
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | - Julie Perroy
- IGF, CNRS, INSERM, Univ Montpellier, Montpellier, France
| | | |
Collapse
|
18
|
Crupi R, Impellizzeri D, Cuzzocrea S. Role of Metabotropic Glutamate Receptors in Neurological Disorders. Front Mol Neurosci 2019; 12:20. [PMID: 30800054 PMCID: PMC6375857 DOI: 10.3389/fnmol.2019.00020] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 01/21/2019] [Indexed: 12/20/2022] Open
Abstract
Glutamate is a fundamental excitatory neurotransmitter in the mammalian central nervous system (CNS), playing key roles in memory, neuronal development, and synaptic plasticity. Moreover, excessive glutamate release has been implicated in neuronal cell death. There are both ionotropic and metabotropic glutamate receptors (mGluRs), the latter of which can be divided into eight subtypes and three subgroups based on homology sequence and their effects on cell signaling. Indeed, mGluRs exert fine control over glutamate activity by stimulating several cell-signaling pathways via the activation of G protein-coupled (GPC) or G protein-independent cell signaling. The involvement of specific mGluRs in different forms of synaptic plasticity suggests that modulation of mGluRs may aid in the treatment of cognitive impairments related to several neurodevelopmental/psychiatric disorders and neurodegenerative diseases, which are associated with a high economic and social burden. Preclinical and clinical data have shown that, in the CNS, mGluRs are able to modulate presynaptic neurotransmission by fine-tuning neuronal firing and neurotransmitter release in a dynamic, activity-dependent manner. Current studies on drugs that target mGluRs have identified promising, innovative pharmacological tools for the treatment of neurodegenerative and neuropsychiatric conditions, including chronic pain.
Collapse
Affiliation(s)
- Rosalia Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy.,Department of Pharmacology and Physiology, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
19
|
Pereira V, Goudet C. Emerging Trends in Pain Modulation by Metabotropic Glutamate Receptors. Front Mol Neurosci 2019; 11:464. [PMID: 30662395 PMCID: PMC6328474 DOI: 10.3389/fnmol.2018.00464] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Pain is an essential protective mechanism meant to prevent tissue damages in organisms. On the other hand, chronic or persistent pain caused, for example, by inflammation or nerve injury is long lasting and responsible for long-term disability in patients. Therefore, chronic pain and its management represents a major public health problem. Hence, it is critical to better understand chronic pain molecular mechanisms to develop innovative and efficient drugs. Over the past decades, accumulating evidence has demonstrated a pivotal role of glutamate in pain sensation and transmission, supporting glutamate receptors as promising potential targets for pain relieving drug development. Glutamate is the most abundant excitatory neurotransmitter in the brain. Once released into the synapse, glutamate acts through ionotropic glutamate receptors (iGluRs), which are ligand-gated ion channels triggering fast excitatory neurotransmission, and metabotropic glutamate receptors (mGluRs), which are G protein-coupled receptors modulating synaptic transmission. Eight mGluRs subtypes have been identified and are divided into three classes based on their sequence similarities and their pharmacological and biochemical properties. Of note, all mGluR subtypes (except mGlu6 receptor) are expressed within the nociceptive pathways where they modulate pain transmission. This review will address the role of mGluRs in acute and persistent pain processing and emerging pharmacotherapies for pain management.
Collapse
Affiliation(s)
- Vanessa Pereira
- IGF, CNRS, INSERM, Univ. de Montpellier, Montpellier, France
| | - Cyril Goudet
- IGF, CNRS, INSERM, Univ. de Montpellier, Montpellier, France
| |
Collapse
|
20
|
Belhocine A, Veglianese P, Hounsou C, Dupuis E, Acher F, Durroux T, Goudet C, Pin JP. Profiling of orthosteric and allosteric group-III metabotropic glutamate receptor ligands on various G protein-coupled receptors with Tag-lite ® assays. Neuropharmacology 2018; 140:233-245. [PMID: 30099051 DOI: 10.1016/j.neuropharm.2018.07.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 12/21/2022]
Abstract
Group-III metabotropic glutamate (mGlu) receptors are important synaptic regulators and are potential druggable targets for Parkinson disease, autism and pain. Potential drugs include orthosteric agonists in the glutamate binding extracellular domain and positive allosteric modulators interacting with seven-pass transmembrane domains. Orthosteric agonists are rarely completely specific for an individual group-III mGlu subtype. Furthermore they often fail to pass the blood-brain barrier and they constitutively activate their target receptor. These properties limit the potential therapeutic use of orthosteric agonists. Allosteric modulators are more specific and maintain the biological activity of the targeted receptor. However, they bind in a hydrophobic pocket and this limits their bio-availability and increases possible off-target action. It is therefore important to characterize the action of potential drug targets with a multifaceted and deeply informative assay. Here we aimed at multifaceted deep profiling of the effect of seven different agonists, and seven positive allosteric modulators on 34 different G protein-coupled receptors by a Tag-lite® assay. Our results did not reveal off-target activity of mGlu orthosteric agonists. However, five allosteric modulators had either positive or negative effects on non-cognate G protein-coupled receptors. In conclusion, we demonstrate the power of the Tag-lite® assay for potential drug ligand profiling on G protein-coupled receptors and its potential to identify positive allosteric compounds.
Collapse
Affiliation(s)
| | | | | | | | - Francine Acher
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Cyril Goudet
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | | |
Collapse
|
21
|
Merighi A. The histology, physiology, neurochemistry and circuitry of the substantia gelatinosa Rolandi (lamina II) in mammalian spinal cord. Prog Neurobiol 2018; 169:91-134. [PMID: 29981393 DOI: 10.1016/j.pneurobio.2018.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 06/07/2018] [Accepted: 06/30/2018] [Indexed: 02/06/2023]
Abstract
The substantia gelatinosa Rolandi (SGR) was first described about two centuries ago. In the following decades an enormous amount of information has permitted us to understand - at least in part - its role in the initial processing of pain and itch. Here, I will first provide a comprehensive picture of the histology, physiology, and neurochemistry of the normal SGR. Then, I will analytically discuss the SGR circuits that have been directly demonstrated or deductively envisaged in the course of the intensive research on this area of the spinal cord, with particular emphasis on the pathways connecting the primary afferent fibers and the intrinsic neurons. The perspective existence of neurochemically-defined sets of primary afferent neurons giving rise to these circuits will be also discussed, with the proposition that a cross-talk between different subsets of peptidergic fibers may be the structural and functional substrate of additional gating mechanisms in SGR. Finally, I highlight the role played by slow acting high molecular weight modulators in these gating mechanisms.
Collapse
Affiliation(s)
- Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095 Grugliasco (TO), Italy.
| |
Collapse
|
22
|
Zussy C, Gómez-Santacana X, Rovira X, De Bundel D, Ferrazzo S, Bosch D, Asede D, Malhaire F, Acher F, Giraldo J, Valjent E, Ehrlich I, Ferraguti F, Pin JP, Llebaria A, Goudet C. Dynamic modulation of inflammatory pain-related affective and sensory symptoms by optical control of amygdala metabotropic glutamate receptor 4. Mol Psychiatry 2018; 23:509-520. [PMID: 27994221 DOI: 10.1038/mp.2016.223] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 10/06/2016] [Accepted: 10/14/2016] [Indexed: 12/30/2022]
Abstract
Contrary to acute pain, chronic pain does not serve as a warning signal and must be considered as a disease per se. This pathology presents a sensory and psychological dimension at the origin of affective and cognitive disorders. Being largely refractory to current pharmacotherapies, identification of endogenous systems involved in persistent and chronic pain is crucial. The amygdala is a key brain region linking pain sensation with negative emotions. Here, we show that activation of a specific intrinsic neuromodulatory system within the amygdala associated with type 4 metabotropic glutamate receptors (mGlu4) abolishes sensory and affective symptoms of persistent pain such as hypersensitivity to pain, anxiety- and depression-related behaviors, and fear extinction impairment. Interestingly, neuroanatomical and synaptic analysis of the amygdala circuitry suggests that the effects of mGlu4 activation occur outside the central nucleus via modulation of multisensory thalamic inputs to lateral amygdala principal neurons and dorso-medial intercalated cells. Furthermore, we developed optogluram, a small diffusible photoswitchable positive allosteric modulator of mGlu4. This ligand allows the control of endogenous mGlu4 activity with light. Using this photopharmacological approach, we rapidly and reversibly inhibited behavioral symptoms associated with persistent pain through optical control of optogluram in the amygdala of freely behaving animals. Altogether, our data identify amygdala mGlu4 signaling as a mechanism that bypasses central sensitization processes to dynamically modulate persistent pain symptoms. Our findings help to define novel and more precise therapeutic interventions for chronic pain, and exemplify the potential of optopharmacology to study the dynamic activity of endogenous neuromodulatory mechanisms in vivo.
Collapse
Affiliation(s)
- C Zussy
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - X Gómez-Santacana
- MCS, Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain.,Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - X Rovira
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - D De Bundel
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - S Ferrazzo
- Department of Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - D Bosch
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - D Asede
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - F Malhaire
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - F Acher
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - J Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Network Biomedical Research Center on Mental Health (CIBERSAM), Madrid, Spain
| | - E Valjent
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - I Ehrlich
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - F Ferraguti
- Department of Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - J-P Pin
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - A Llebaria
- MCS, Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - C Goudet
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| |
Collapse
|
23
|
Volpi C, Fallarino F, Mondanelli G, Macchiarulo A, Grohmann U. Opportunities and challenges in drug discovery targeting metabotropic glutamate receptor 4. Expert Opin Drug Discov 2018; 13:411-423. [DOI: 10.1080/17460441.2018.1443076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Claudia Volpi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
24
|
Selvam C, Lemasson IA, Brabet I, Oueslati N, Karaman B, Cabaye A, Tora AS, Commare B, Courtiol T, Cesarini S, McCort-Tranchepain I, Rigault D, Mony L, Bessiron T, McLean H, Leroux FR, Colobert F, Daniel H, Goupil-Lamy A, Bertrand HO, Goudet C, Pin JP, Acher FC. Increased Potency and Selectivity for Group III Metabotropic Glutamate Receptor Agonists Binding at Dual sites. J Med Chem 2018; 61:1969-1989. [DOI: 10.1021/acs.jmedchem.7b01438] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Chelliah Selvam
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Isabelle A. Lemasson
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Isabelle Brabet
- IGF, CNRS, INSERM, Université Montpellier, F-34094 Montpellier, France
| | - Nadia Oueslati
- IGF, CNRS, INSERM, Université Montpellier, F-34094 Montpellier, France
| | - Berin Karaman
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Alexandre Cabaye
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Amélie S. Tora
- IGF, CNRS, INSERM, Université Montpellier, F-34094 Montpellier, France
| | - Bruno Commare
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
- UMR 7509/CNRS/ECPM, Université de Strasbourg, 25 Rue Becquerel, 67087 Strasbourg 02, France
| | - Tiphanie Courtiol
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Sara Cesarini
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Isabelle McCort-Tranchepain
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Delphine Rigault
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Laetitia Mony
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
- Institut de Biologie, Ecole Normale Supérieure, CNRS UMR 8197, INSERM U1024, PSL University, 46 rue d’Ulm, 75005 Paris, France
| | - Thomas Bessiron
- Pharmacologie et Biochimie de la Synapse, Université Paris-Sud/CNRS/NeuroPSI−UMR 9197, F-91405 Orsay, France
| | - Heather McLean
- Pharmacologie et Biochimie de la Synapse, Université Paris-Sud/CNRS/NeuroPSI−UMR 9197, F-91405 Orsay, France
| | - Frédéric R. Leroux
- UMR 7509/CNRS/ECPM, Université de Strasbourg, 25 Rue Becquerel, 67087 Strasbourg 02, France
| | - Françoise Colobert
- UMR 7509/CNRS/ECPM, Université de Strasbourg, 25 Rue Becquerel, 67087 Strasbourg 02, France
| | - Hervé Daniel
- Pharmacologie et Biochimie de la Synapse, Université Paris-Sud/CNRS/NeuroPSI−UMR 9197, F-91405 Orsay, France
| | - Anne Goupil-Lamy
- BIOVIA, Dassault Systèmes, 10 rue Marcel Dassault, CS 40501, 78946 Vélizy-Villacoublay Cedex, France
| | - Hugues-Olivier Bertrand
- BIOVIA, Dassault Systèmes, 10 rue Marcel Dassault, CS 40501, 78946 Vélizy-Villacoublay Cedex, France
| | - Cyril Goudet
- IGF, CNRS, INSERM, Université Montpellier, F-34094 Montpellier, France
| | - Jean-Philippe Pin
- IGF, CNRS, INSERM, Université Montpellier, F-34094 Montpellier, France
| | - Francine C. Acher
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| |
Collapse
|
25
|
Hajasova Z, Canestrelli C, Acher F, Noble F, Marie N. Role of mGlu7 receptor in morphine rewarding effects is uncovered by a novel orthosteric agonist. Neuropharmacology 2018; 131:424-430. [PMID: 29307544 DOI: 10.1016/j.neuropharm.2018.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 12/26/2017] [Accepted: 01/02/2018] [Indexed: 11/26/2022]
Abstract
Opiate dependence is a major health issue and despite the existence of opioid substitution treatment, relapse frequently occurs. Group III metabotropic glutamate (mGlu) receptors has received much attention as a putative target in ethanol and cocaine addiction, but no data on opiate addiction exist. So we investigated the role of group III mGlu receptors in morphine rewarding effects through the expression and the reinstatement of conditioned place preference (CPP) using a newly synthesized mGlu4/mGlu7 receptor orthosteric agonist, LSP2-9166. We found that LSP2-9166 blocked morphine CPP expression and reinstatement after extinction. Blockade of CPP expression with LSP2-9166 was abolished when using XAP044, a mGlu7 antagonist. We also found that LSP2-9166 at the dose active for blocking morphine reward was devoid of any effect on locomotion, hedonic state, spatial memory, anxiety or depression. Altogether our data demonstrated that group III mGlu receptors, and more specifically mGlu7, might be a valuable target in opiate addiction.
Collapse
Affiliation(s)
- Zuzana Hajasova
- CNRS ERL 3649, Neuroplasticité et Thérapie des Addictions, Paris, France; INSERM UMR-S 1124, Paris, France; Université Paris Descartes, Paris, France
| | - Corinne Canestrelli
- CNRS ERL 3649, Neuroplasticité et Thérapie des Addictions, Paris, France; INSERM UMR-S 1124, Paris, France; Université Paris Descartes, Paris, France
| | - Francine Acher
- Université Paris Descartes, Paris, France; CNRS UMR8601, Laboratoire de Chimie & Biochimie Pharmacologiques et Toxicologiques, Paris, France
| | - Florence Noble
- CNRS ERL 3649, Neuroplasticité et Thérapie des Addictions, Paris, France; INSERM UMR-S 1124, Paris, France; Université Paris Descartes, Paris, France
| | - Nicolas Marie
- CNRS ERL 3649, Neuroplasticité et Thérapie des Addictions, Paris, France; INSERM UMR-S 1124, Paris, France; Université Paris Descartes, Paris, France.
| |
Collapse
|
26
|
Yasko JR, Mains RE. Chronic pain following spinal cord injury: Current approaches to cellular and molecular mechanisms. TRENDS IN CELL & MOLECULAR BIOLOGY 2018; 13:67-84. [PMID: 33564218 PMCID: PMC7869846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Traumatic spinal cord injury (SCI) has devastating implications for patients, including a high prevalence of chronic pain. Despite advancements in our understanding of the mechanisms involved post-SCI, there are no effective treatments for chronic pain following injury. The development of new treatment interventions for pain is needed, but this requires improved models to assess injury-related cellular, neurophysiological and molecular changes in the spinal cord. Here, we will discuss recent animal models for SCI, molecular screening for altered patterns of gene expression, and the importance of injury severity and timing after SCI.
Collapse
Affiliation(s)
- Jessica R. Yasko
- Department of Neuroscience, University of Connecticut Health Center, Farmington CT 06030-3401, USA
| | - Richard E. Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington CT 06030-3401, USA
| |
Collapse
|
27
|
Chagas PM, da Cruz Weber Fulco B, Pesarico AP, Roehrs JA, Nogueira CW. Effectiveness of bis(phenylimidazoselenazolyl) diselenide on a mouse model of inflammatory nociception. Biomed Pharmacother 2017; 96:56-63. [PMID: 28963951 DOI: 10.1016/j.biopha.2017.09.119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/11/2017] [Accepted: 09/23/2017] [Indexed: 11/16/2022] Open
Abstract
The injection of complete Freund's adjuvant (CFA) in the hindpaw of rodents induces tissue inflammation and nociceptive hypersensitivity. In addition, it has been reported that organoselenium compounds have antinociceptive properties in animal models. The purpose of this study was to investigate the potential antinociceptive effect of bis(phenylimidazoselenazolyl) diselenide (BPIS) in the inflammatory nociception model in mice and its possible mechanism of action. C57BL/6 mice received CFA intraplantar in right hindpaw and the inflammatory response was verified 24h after injection as well as the antinociceptive effect of BPIS. The CFA-induced mechanical allodynia was reversed by BPIS treatment (1mg/kg, p.o.) observed through the von Frey hair test. Additionally, L-arginine (600mg/kg; i.p.), administered before BPIS treatment, blocked its antinociceptive effect. Regarding myeloperoxidase activity, NOx and 3-nitrotyrosine levels, BPIS administration did not reverse alterations observed in the paw of animals injected with CFA. BPIS reversed the increase in spinal NOx content induced by CFA. In the spinal cord, it was also found that CFA induced an increase in malondialdehyde content and a decrease in glutamate uptake, and these alterations were reversed by BPIS. Moreover, BPIS treatment induced an increase in non-protein thiol levels in spinal cord of animals that received CFA injection. No alterations were found in glutathione peroxidase, reductase and S-transferase activities of experimental groups. The obtained data reinforce the relevance of BPIS as an antinociceptive agent as well as highlight the importance of the nitric oxide pathway in the spinal cord and its antioxidant potential for its mechanism of action.
Collapse
Affiliation(s)
- Pietro Maria Chagas
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil
| | - Bruna da Cruz Weber Fulco
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil
| | - Ana Paula Pesarico
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil
| | - Juliano Alex Roehrs
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil
| | - Cristina Wayne Nogueira
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil.
| |
Collapse
|
28
|
Lian YN, Lu Q, Chang JL, Zhang Y. The role of glutamate and its receptors in central nervous system in stress-induced hyperalgesia. Int J Neurosci 2017; 128:283-290. [DOI: 10.1080/00207454.2017.1387112] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yan-Na Lian
- Department of Physiology, Harbin Medical University, Harbin, P. R. China
| | - Qi Lu
- Department of Physiology, Harbin Medical University, Harbin, P. R. China
| | - Jin-Long Chang
- Department of Physiology, Harbin Medical University, Harbin, P. R. China
| | - Ying Zhang
- Department of Physiology, Harbin Medical University, Harbin, P. R. China
| |
Collapse
|
29
|
Dalton JAR, Pin JP, Giraldo J. Analysis of positive and negative allosteric modulation in metabotropic glutamate receptors 4 and 5 with a dual ligand. Sci Rep 2017; 7:4944. [PMID: 28694498 PMCID: PMC5504000 DOI: 10.1038/s41598-017-05095-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/25/2017] [Indexed: 12/24/2022] Open
Abstract
As class C GPCRs and regulators of synaptic activity, human metabotropic glutamate receptors (mGluRs) 4 and 5 are prime targets for allosteric modulation, with mGlu5 inhibition or mGlu4 stimulation potentially treating conditions like chronic pain and Parkinson’s disease. As an allosteric modulator that can bind both receptors, 2-Methyl-6-(phenylethynyl)pyridine (MPEP) is able to negatively modulate mGlu5 or positively modulate mGlu4. At a structural level, how it elicits these responses and how mGluRs undergo activation is unclear. Here, we employ homology modelling and 30 µs of atomistic molecular dynamics (MD) simulations to probe allosteric conformational change in mGlu4 and mGlu5, with and without docked MPEP. Our results identify several structural differences between mGlu4 and mGlu5, as well as key differences responsible for MPEP-mediated positive and negative allosteric modulation, respectively. A novel mechanism of mGlu4 activation is revealed, which may apply to all mGluRs in general. This involves conformational changes in TM3, TM4 and TM5, separation of intracellular loop 2 (ICL2) from ICL1/ICL3, and destabilization of the ionic-lock. On the other hand, mGlu5 experiences little disturbance when MPEP binds, maintaining its inactive state with reduced conformational fluctuation. In addition, when MPEP is absent, a lipid molecule can enter the mGlu5 allosteric pocket.
Collapse
Affiliation(s)
- James A R Dalton
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Network Biomedical Research Centre on Mental Health (CIBERSAM), Madrid, Spain
| | - Jean-Philippe Pin
- Institute of Functional Genomics, Université de Montpellier, Unité Mixte de Recherche 5302 CNRS, Montpellier, France.,Unité de recherche U1191, INSERM, Montpellier, France
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain. .,Network Biomedical Research Centre on Mental Health (CIBERSAM), Madrid, Spain.
| |
Collapse
|
30
|
Chagas PM, Fulco BCW, Sari MHM, Roehrs JA, Nogueira CW. Bis(phenylimidazoselenazolyl) diselenide elicits antinociceptive effect by modulating myeloperoxidase activity, NOx and NFkB levels in the collagen-induced arthritis mouse model. ACTA ACUST UNITED AC 2017; 69:1022-1032. [PMID: 28436532 DOI: 10.1111/jphp.12738] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/26/2017] [Indexed: 12/15/2022]
Abstract
Bis(phenylimidazoselenazolyl) diselenide (BPIS) is an organoselenium with acute antinociceptive and antioxidant properties. OBJECTIVES The aim of this study was to investigate BPIS effect on a collagen-induced arthritis (CIA) model in mice. METHODS Protocol of exposure consisted in arthritis induction by chicken collagen type II on day 0 with booster injection on day 21. On day 60 after collagen injection, incidence of mechanic allodynia (Von Frey test) or thermal hyperalgesia (hot plate test) was evaluated. During following 5 days, mice were treated with BPIS (0.1-1 mg/kg; p.o.; daily) or vehicle. On day 65, mice were killed, and paws and spinal cord were removed for analyses. KEY FINDINGS Mice submitted to CIA model developed both mechanical allodynia and thermal hyperalgesia, which were reversed by BPIS at the highest dose. In paw, BPIS reversed the increase in myeloperoxidase activity in the CIA group. In the spinal cord, BPIS decreased NOx and NFkB levels increased in the CIA group. BPIS-treated animals had lower cyclooxygenase-2 levels in the spinal cord. CONCLUSIONS The myeloperoxidase activity in paw and NOx and NFkB levels in spinal cord are related to antinociceptive properties of BPIS in CIA model.
Collapse
Affiliation(s)
- Pietro M Chagas
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Bruna C W Fulco
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Marcel H M Sari
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Juliano A Roehrs
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.,Departamento de Química, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Cristina W Nogueira
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
31
|
Rovira X, Trapero A, Pittolo S, Zussy C, Faucherre A, Jopling C, Giraldo J, Pin JP, Gorostiza P, Goudet C, Llebaria A. OptoGluNAM4.1, a Photoswitchable Allosteric Antagonist for Real-Time Control of mGlu4 Receptor Activity. Cell Chem Biol 2016; 23:929-34. [PMID: 27478159 DOI: 10.1016/j.chembiol.2016.06.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 11/17/2022]
Abstract
OptoGluNAM4.1, a negative allosteric modulator (NAM) of metabotropic glutamate receptor 4 (mGlu4) contains a reactive group that covalently binds to the receptor and a blue-light-activated, fast-relaxing azobenzene group that allows reversible receptor activity photocontrol in vitro and in vivo. OptoGluNAM4.1 induces light-dependent behavior in zebrafish and reverses the activity of the mGlu4 agonist LSP4-2022 in a mice model of chronic pain, defining a photopharmacological tool to better elucidate the physiological roles of the mGlu4 receptor in the nervous system.
Collapse
Affiliation(s)
- Xavier Rovira
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, 34000 Montpellier, France; INSERM, U1191, 34000 Montpellier, France
| | - Ana Trapero
- Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain; Institute for Bioengineering of Catalonia (IBEC), 08028 Barcelona, Spain
| | - Silvia Pittolo
- Institute for Bioengineering of Catalonia (IBEC), 08028 Barcelona, Spain
| | - Charleine Zussy
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, 34000 Montpellier, France; INSERM, U1191, 34000 Montpellier, France
| | - Adèle Faucherre
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, 34000 Montpellier, France; INSERM, U1191, 34000 Montpellier, France
| | - Chris Jopling
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, 34000 Montpellier, France; INSERM, U1191, 34000 Montpellier, France
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, 34000 Montpellier, France; INSERM, U1191, 34000 Montpellier, France
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), 08028 Barcelona, Spain; Network Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Poeta Mariano Esquillor s/n, Zaragoza 50018, Spain; ICREA, Pg. Lluís Companys, 23, Barcelona 08010, Spain.
| | - Cyril Goudet
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, 34000 Montpellier, France; INSERM, U1191, 34000 Montpellier, France.
| | - Amadeu Llebaria
- Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain.
| |
Collapse
|
32
|
Chiechio S. Modulation of Chronic Pain by Metabotropic Glutamate Receptors. PHARMACOLOGICAL MECHANISMS AND THE MODULATION OF PAIN 2016; 75:63-89. [DOI: 10.1016/bs.apha.2015.11.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
33
|
Yu J, Ding CP, Wang J, Wang T, Zhang T, Zeng XY, Wang JY. Red nucleus glutamate facilitates neuropathic allodynia induced by spared nerve injury through non-NMDA and metabotropic glutamate receptors. J Neurosci Res 2015; 93:1839-48. [DOI: 10.1002/jnr.23671] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 08/31/2015] [Accepted: 08/31/2015] [Indexed: 01/23/2023]
Affiliation(s)
- Jing Yu
- Department of Immunology and Pathogenic Biology; Xi'an Jiaotong University Health Science Center; Xi'an Shaanxi People's Republic of China
| | - Cui-Ping Ding
- Department of Immunology and Pathogenic Biology; Xi'an Jiaotong University Health Science Center; Xi'an Shaanxi People's Republic of China
| | - Jing Wang
- Department of Immunology and Pathogenic Biology; Xi'an Jiaotong University Health Science Center; Xi'an Shaanxi People's Republic of China
| | - Ting Wang
- Department of Immunology and Pathogenic Biology; Xi'an Jiaotong University Health Science Center; Xi'an Shaanxi People's Republic of China
- Department of Nuclear Medicine; Ankang City Center Hospital; Ankang Shaanxi People's Republic of China
| | - Tao Zhang
- Department of Immunology and Pathogenic Biology; Xi'an Jiaotong University Health Science Center; Xi'an Shaanxi People's Republic of China
- Department of Nuclear Medicine; Ankang City Center Hospital; Ankang Shaanxi People's Republic of China
| | - Xiao-Yan Zeng
- Department of Laboratory Medicine, The First Affiliated Hospital; Xi'an Jiaotong University Health Science Center; Xi'an Shaanxi People's Republic of China
| | - Jun-Yang Wang
- Department of Immunology and Pathogenic Biology; Xi'an Jiaotong University Health Science Center; Xi'an Shaanxi People's Republic of China
| |
Collapse
|
34
|
Podkowa K, Rzeźniczek S, Marciniak M, Acher F, Pilc A, Pałucha-Poniewiera A. A novel mGlu4 selective agonist LSP4-2022 increases behavioral despair in mouse models of antidepressant action. Neuropharmacology 2015; 97:338-45. [PMID: 26074092 DOI: 10.1016/j.neuropharm.2015.05.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/26/2015] [Accepted: 05/30/2015] [Indexed: 11/16/2022]
Abstract
Numerous data have indicated that metabotropic glutamate (mGlu) receptor ligands may be potentially useful as novel antidepressant drugs (ADs). The Group III mGlu receptor has not been explored much because of the limited access to selective ligands, but some behavioral studies have indicated that modulating group III mGlu receptors may result in benefits for the therapy of depression. Here, we investigated the potential antidepressant-like effects of a new mGlu4 selective orthosteric agonist, LSP4-2022. We found that the drug induced pro-depressant effects in the tail suspension test (TST) and the forced swim test (FST) in mice at doses that did not change the locomotor activity of the animals. Additional experiments that used knock-out (KO) mice and aimed to verify the selectivity of LSP4-2022 revealed that the drug induced strong pro-depressant-like effects in mGlu7 KO mice but did not affect the behavior of mGlu4 KO mice in the TST, suggesting that the activation of the mGlu4 receptor plays a role in producing the pro-depressant activity of the tested drug. The results of our study indicate that the inhibition rather than activation of mGlu4 receptors might induce antidepressant effects, but this hypothesis should be verified using a selective mGlu4 receptor antagonist, which is currently not available.
Collapse
Affiliation(s)
- Karolina Podkowa
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland
| | - Szymon Rzeźniczek
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland
| | - Marcin Marciniak
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland
| | - Francine Acher
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, UMR8601-CNRS, Paris Descartes University, Sorbonne Paris Cite,45, rue des Saints-Peres, 75270 Paris Cedex 06, France
| | - Andrzej Pilc
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland; Jagiellonian University Medical College, Department of Drug Management, Faculty of Health Sciences, Grzegórzecka 20, 31-531 Kraków, Poland
| | - Agnieszka Pałucha-Poniewiera
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland.
| |
Collapse
|
35
|
Gregory KJ, Conn PJ. Molecular Insights into Metabotropic Glutamate Receptor Allosteric Modulation. Mol Pharmacol 2015; 88:188-202. [PMID: 25808929 DOI: 10.1124/mol.114.097220] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/24/2015] [Indexed: 12/21/2022] Open
Abstract
The metabotropic glutamate (mGlu) receptors are a group of eight family C G protein-coupled receptors that are expressed throughout the central nervous system (CNS) and periphery. Within the CNS the different subtypes are found in neurons, both pre- and/or postsynaptically, where they mediate modulatory roles and in glial cells. The mGlu receptor family provides attractive targets for numerous psychiatric and neurologic disorders, with the majority of discovery programs focused on targeting allosteric sites, with allosteric ligands now available for all mGlu receptor subtypes. However, the development of allosteric ligands remains challenging. Biased modulation, probe dependence, and molecular switches all contribute to the complex molecular pharmacology exhibited by mGlu receptor allosteric ligands. In recent years we have made significant progress in our understanding of this molecular complexity coupled with an increased understanding of the structural basis of mGlu allosteric modulation.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Vanderbilt Center for Neuroscience Drug Discovery & Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (P.J.C)
| | - P Jeffrey Conn
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Vanderbilt Center for Neuroscience Drug Discovery & Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (P.J.C)
| |
Collapse
|
36
|
Therapeutic potential of group III metabotropic glutamate receptor ligands in pain. Curr Opin Pharmacol 2015; 20:64-72. [DOI: 10.1016/j.coph.2014.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/18/2014] [Accepted: 11/21/2014] [Indexed: 11/22/2022]
|
37
|
Johnson AC, Greenwood-Van Meerveld B. Stress-induced pain: a target for the development of novel therapeutics. J Pharmacol Exp Ther 2014; 351:327-35. [PMID: 25194019 PMCID: PMC4201269 DOI: 10.1124/jpet.114.218065] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/04/2014] [Indexed: 12/12/2022] Open
Abstract
Although current therapeutics provide relief from acute pain, drugs used for treatment of chronic pain are typically less efficacious and limited by adverse side effects, including tolerance, addiction, and gastrointestinal upset. Thus, there is a significant need for novel therapies for the treatment of chronic pain. In concert with chronic pain, persistent stress facilitates pain perception and sensitizes pain pathways, leading to a feed-forward cycle promoting chronic pain disorders. Stress exacerbation of chronic pain suggests that centrally acting drugs targeting the pain- and stress-responsive brain regions represent a valid target for the development of novel therapeutics. This review provides an overview of how stress modulates spinal and central pain pathways, identifies key neurotransmitters and receptors within these pathways, and highlights their potential as novel targets for therapeutics to treat chronic pain.
Collapse
Affiliation(s)
- Anthony C Johnson
- Veterans Affairs Medical Center (B.G.-V.M.), Department of Physiology (B.G.-V.M.), and Oklahoma Center for Neuroscience (A.C.J., B.G.-V.M.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Beverley Greenwood-Van Meerveld
- Veterans Affairs Medical Center (B.G.-V.M.), Department of Physiology (B.G.-V.M.), and Oklahoma Center for Neuroscience (A.C.J., B.G.-V.M.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
38
|
Rovira X, Malhaire F, Scholler P, Rodrigo J, Gonzalez-Bulnes P, Llebaria A, Pin JP, Giraldo J, Goudet C. Overlapping binding sites drive allosteric agonism and positive cooperativity in type 4 metabotropic glutamate receptors. FASEB J 2014; 29:116-30. [PMID: 25342125 DOI: 10.1096/fj.14-257287] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Type 4 metabotropic glutamate (mGlu4) receptors are emerging targets for the treatment of various disorders. Accordingly, numerous mGlu4-positive allosteric modulators (PAMs) have been identified, some of which also display agonist activity. To identify the structural bases for their allosteric action, we explored the relationship between the binding pockets of mGlu4 PAMs with different chemical scaffolds and their functional properties. By use of innovative mGlu4 biosensors and second-messenger assays, we show that all PAMs enhance agonist action on the receptor through different degrees of allosteric agonism and positive cooperativity. For example, whereas VU0155041 and VU0415374 display equivalent efficacies [log(τ(B)) = 1.15 ± 0.38 and 1.25 ± 0.44, respectively], they increase the ability of L-AP4 to stabilize the active conformation of the receptor by 4 and 39 times, respectively. Modeling and docking studies identify 2 overlapping binding pockets as follows: a first site homologous to the pocket of natural agonists of class A GPCRs linked to allosteric agonism and a second one pointing toward a site topographically homologous to the Na(+) binding pocket of class A GPCRs, occupied by PAMs exhibiting the strongest cooperativity. These results reveal that intrinsic efficacy and cooperativity of mGlu4 PAMs are correlated with their binding mode, and vice versa, integrating structural and functional knowledge from different GPCR classes.
Collapse
Affiliation(s)
- Xavier Rovira
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, Université de Montpellier, Montpellier, France; INSERM, U661, Montpellier, France
| | - Fanny Malhaire
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, Université de Montpellier, Montpellier, France; INSERM, U661, Montpellier, France
| | - Pauline Scholler
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, Université de Montpellier, Montpellier, France; INSERM, U661, Montpellier, France
| | - Jordi Rodrigo
- Laboratoire de Chimie Thérapeutique, BioCIS UMR-CNRS 8076, LabEx LERMIT, Faculté de Pharmacie, Université Paris-Sud, Châtenay-Malabry, Paris, France
| | - Patricia Gonzalez-Bulnes
- Laboratory of Medicinal Chemistry, Departament of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia IQAC-CSIC, Barcelona, Spain; and
| | - Amadeu Llebaria
- Laboratory of Medicinal Chemistry, Departament of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia IQAC-CSIC, Barcelona, Spain; and
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, Université de Montpellier, Montpellier, France; INSERM, U661, Montpellier, France
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Cyril Goudet
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, Université de Montpellier, Montpellier, France; INSERM, U661, Montpellier, France;
| |
Collapse
|
39
|
Gaillard S, Lo Re L, Mantilleri A, Hepp R, Urien L, Malapert P, Alonso S, Deage M, Kambrun C, Landry M, Low SA, Alloui A, Lambolez B, Scherrer G, Le Feuvre Y, Bourinet E, Moqrich A. GINIP, a Gαi-interacting protein, functions as a key modulator of peripheral GABAB receptor-mediated analgesia. Neuron 2014; 84:123-136. [PMID: 25242222 DOI: 10.1016/j.neuron.2014.08.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2014] [Indexed: 12/15/2022]
Abstract
One feature of neuropathic pain is a reduced GABAergic inhibitory function. Nociceptors have been suggested to play a key role in this process. However, the mechanisms behind nociceptor-mediated modulation of GABA signaling remain to be elucidated. Here we describe the identification of GINIP, a Gαi-interacting protein expressed in two distinct subsets of nonpeptidergic nociceptors. GINIP null mice develop a selective and prolonged mechanical hypersensitivity in models of inflammation and neuropathy. GINIP null mice show impaired responsiveness to GABAB, but not to delta or mu opioid receptor agonist-mediated analgesia specifically in the spared nerve injury (SNI) model. Consistently, GINIP-deficient dorsal root ganglia neurons had lower baclofen-evoked inhibition of high-voltage-activated calcium channels and a defective presynaptic inhibition of lamina IIi interneurons. These results further support the role of unmyelinated C fibers in injury-induced modulation of spinal GABAergic inhibition and identify GINIP as a key modulator of peripherally evoked GABAB-receptors signaling.
Collapse
Affiliation(s)
- Stéphane Gaillard
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, France
| | - Laure Lo Re
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, France
| | - Annabelle Mantilleri
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, France
| | - Régine Hepp
- Sorbonne Universités, UPMC Univ Paris 06, UM CR 18, Neuroscience Paris Seine, 75005 Paris, France; Centre National de la Recherche Scientifique (CNRS), UMR 8246 Paris, France; Institut national de la Santé et de la Recherche Médicale (INSERM), UMR-S 1130 Paris, France
| | - Louise Urien
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, France
| | - Pascale Malapert
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, France
| | - Serge Alonso
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, France
| | - Michael Deage
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, UMR 5203, CNRS, U661, INSERM, Universités Montpellier I&II, 141 Rue de la Cardonille, 34094 Montpellier Cedex 05, France
| | - Charline Kambrun
- University Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France
| | - Marc Landry
- University Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France
| | - Sarah A Low
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Abdelkrim Alloui
- Laboratoire de Pharmacologie Médicale, Faculté de Médecine et de Pharmacie, UMR 766 INSERM, 28 place Henri-Dunant, BP 38, 63001 Clermont-Ferrand Cedex 1, France
| | - Bertrand Lambolez
- Sorbonne Universités, UPMC Univ Paris 06, UM CR 18, Neuroscience Paris Seine, 75005 Paris, France; Centre National de la Recherche Scientifique (CNRS), UMR 8246 Paris, France; Institut national de la Santé et de la Recherche Médicale (INSERM), UMR-S 1130 Paris, France
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Yves Le Feuvre
- University Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France
| | - Emmanuel Bourinet
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, UMR 5203, CNRS, U661, INSERM, Universités Montpellier I&II, 141 Rue de la Cardonille, 34094 Montpellier Cedex 05, France
| | - Aziz Moqrich
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, France.
| |
Collapse
|
40
|
Rovira X, Harrak Y, Trapero A, González-Bulnes P, Malhaire F, Pin JP, Goudet C, Giraldo J, Llebaria A. Exploring the active conformation of cyclohexane carboxylate positive allosteric modulators of the type 4 metabotropic glutamate receptor. ChemMedChem 2014; 9:2685-98. [PMID: 25196639 DOI: 10.1002/cmdc.201402190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Indexed: 11/07/2022]
Abstract
The active conformation of a family of metabotropic glutamate receptor subtype 4 (mGlu4 ) positive allosteric modulators (PAMs) with the cyclohexane 1,2-dicarboxylic scaffold present in cis-2-(3,5-dichlorophenylcarbamoyl)cyclohexanecarboxylic acid (VU0155041) was investigated by testing structurally similar six-membered ring compounds that have a locked conformation. The norbornane and cyclohexane molecules designed as mGlu4 conformational probes and the enantiomers of the trans diastereomer were computationally characterized and tested in mGlu4 pharmacological assays. The results support a VU0155041 active conformation, with the chair cyclohexane having the aromatic amide substituent in an axial position and the carboxylate in an equatorial position. Moreover, the receptor displays enantiomeric discrimination of the chiral PAMs. The constructed pharmacophore characterized a highly constrained mGlu4 allosteric binding site, thus providing a step forward in structure-based drug design for mGlu4 PAMs.
Collapse
Affiliation(s)
- Xavier Rovira
- Laboratory of Molecular Neuropharmacology & Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, 08193 Bellaterra (Spain); Institut de Génomique Fonctionnelle, CNRS UMR5203, Université de Montpellier; U661, INSERM, 141 Rue de la Cardonille, 34094 Montpellier (France)
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Gómez-Santacana X, Rovira X, Dalton JA, Goudet C, Pin JP, Gorostiza P, Giraldo J, Llebaria A. A double effect molecular switch leads to a novel potent negative allosteric modulator of metabotropic glutamate receptor 5. MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00208c] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
42
|
Golubeva AV, Zhdanov AV, Mallel G, Dinan TG, Cryan JF. The mouse cyclophosphamide model of bladder pain syndrome: tissue characterization, immune profiling, and relationship to metabotropic glutamate receptors. Physiol Rep 2014; 2:e00260. [PMID: 24760514 PMCID: PMC4002240 DOI: 10.1002/phy2.260] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 02/17/2014] [Indexed: 01/08/2023] Open
Abstract
Abstract Painful bladder syndrome/Interstitial cystitis (PBS/IC) is a chronic disorder characterized clinically by recurring episodes of pelvic pain and increased urination frequency, significantly impairing patients' quality of life. Despite this, there is an unmet medical need in terms of effective diagnostics and treatment. Animal models are crucial in this endeavor. Systemic chronic administration of cyclophosphamide (CYP) in mice has been proposed as a relevant preclinical model of chronic bladder pain. However, molecular mechanisms underlying the pathogenesis of this model are lacking. Here, we show that mice, subjected to repetitive systemic injections of CYP, developed mild inflammatory response in bladder tissue characterized by submucosal edema, moderate increase in proinflammatory cytokine gene expression, and mastocytosis. No signs of massive inflammatory infiltrate, tissue hemorrhages, mucosal ulcerations and urothelium loss were observed. Instead, CYP treatment induced urothelium hyperplasia, accompanied by activation of proliferative signaling cascades, and a decrease in the expression of urothelium-specific markers. Metabotropic glutamate (mGlu) receptors have been implicated in chronic pain disorders. CYP administration induced differential changes in mGlu receptors mRNA levels in bladder tissue, without affecting gene expression at spinal cord level, pointing to the potential link between peripheral mGlu receptors and inflammation-induced bladder malfunction and hyperalgesia. Taken together, these data indicate that chronic CYP treatment in mice is a model of PBS mostly relevant to the major, nonulcerative subtype of the syndrome, characterized by a relatively unaltered mucosa and a sparse inflammatory response. This model can help to elucidate the pathogenetic mechanisms of the disease.
Collapse
Affiliation(s)
- Anna V. Golubeva
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | | | - Giuseppe Mallel
- Pathology Unit, Department of Clinical and Molecular Medicine, S. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Timothy G. Dinan
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
- Department of Psychiatry, University College Cork, Cork, Ireland
| | - John F. Cryan
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|