1
|
Domin H, Burnat G. mGlu4R, mGlu7R, and mGlu8R allosteric modulation for treating acute and chronic neurodegenerative disorders. Pharmacol Rep 2024; 76:1219-1241. [PMID: 39348087 PMCID: PMC11582148 DOI: 10.1007/s43440-024-00657-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024]
Abstract
Neuroprotection, defined as safeguarding neurons from damage and death by inhibiting diverse pathological mechanisms, continues to be a promising approach for managing a range of central nervous system (CNS) disorders, including acute conditions such as ischemic stroke and traumatic brain injury (TBI) and chronic neurodegenerative diseases like Parkinson's disease (PD), Alzheimer's disease (AD), and multiple sclerosis (MS). These pathophysiological conditions involve excessive glutamatergic (Glu) transmission activity, which can lead to excitotoxicity. Inhibiting this excessive Glu transmission has been proposed as a potential therapeutic strategy for treating the CNS disorders mentioned. In particular, ligands of G protein-coupled receptors (GPCRs), including metabotropic glutamatergic receptors (mGluRs), have been recognized as promising options for inhibiting excessive Glu transmission. This review discusses the complex interactions of mGlu receptors with their subtypes, including the formation of homo- and heterodimers, which may vary in function and pharmacology depending on their protomer composition. Understanding these intricate details of mGlu receptor structure and function enhances researchers' ability to develop targeted pharmacological interventions, potentially offering new therapeutic avenues for neurological and psychiatric disorders. This review also summarizes the current knowledge of the neuroprotective potential of ligands targeting group III mGluRs in preclinical cellular (in vitro) and animal (in vivo) models of ischemic stroke, TBI, PD, AD, and MS. In recent years, experiments have shown that compounds, especially those activating mGlu4 or mGlu7 receptors, exhibit protective effects in experimental ischemia models. The discovery of allosteric ligands for specific mGluR subtypes has led to reports suggesting that group III mGluRs may be promising targets for neuroprotective therapy in PD (mGlu4R), TBI (mGlu7R), and MS (mGlu8R).
Collapse
Affiliation(s)
- Helena Domin
- Maj Institute of Pharmacology, Department of Neurobiology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland.
| | - Grzegorz Burnat
- Maj Institute of Pharmacology, Department of Neurobiology, Polish Academy of Sciences, Smętna 12, Kraków, 31-343, Poland
| |
Collapse
|
2
|
Li P, Lei W, Dong Y, Wang X, Ye X, Tian Y, Yang Y, Liu J, Li N, Niu X, Wang X, Tian Y, Xu L, Yang Y, Liu J. mGluR7: The new player protecting the central nervous system. Ageing Res Rev 2024; 102:102554. [PMID: 39454762 DOI: 10.1016/j.arr.2024.102554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
Metabotropic glutamate receptor 7 (mGluR7) belongs to the family of type III mGluR receptor, playing an important part in the central nervous system (CNS) through response to neurotransmitter regulation, reduction of excitatory toxicity, and early neuronal development. Drugs targeting mGluR7 (mGluR7 agonists, antagonists, and allosteric modulators) may be among the most promising agents for the treatment of CNS disorders, such as psychiatric disorders, neurodegenerative diseases, and neurodevelopmental impairments, though these potential therapies are at early stages and the data are still limited. In this review, we summarized the structure and function of mGluR7 and discussed recent progress on mGluR7 agonists and antagonists. A deeper understanding of mGluR7 will contribute to uncovering the molecular mechanisms of neuroprotection and providing a theoretical basis for the formulation of therapeutic strategies.
Collapse
Affiliation(s)
- Pan Li
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Ophthalmology, Xi'an No.1 Hospital, Faculty of Life Sciences and Medicine, Northwest University, 30 Fenxiang Road, Xi'an 710002, China
| | - Wangrui Lei
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yushu Dong
- Department of Neurosurgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Xingyan Ye
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yaru Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Jie Liu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Ning Li
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xiaochen Niu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xin Wang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yifan Tian
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Lu Xu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| | - Jincheng Liu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
3
|
Han S, Wang J, Zhang W, Tian X. Chronic Pain-Related Cognitive Deficits: Preclinical Insights into Molecular, Cellular, and Circuit Mechanisms. Mol Neurobiol 2024; 61:8123-8143. [PMID: 38470516 DOI: 10.1007/s12035-024-04073-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/23/2024] [Indexed: 03/14/2024]
Abstract
Cognitive impairment is a common comorbidity of chronic pain, significantly disrupting patients' quality of life. Despite this comorbidity being clinically recognized, the underlying neuropathological mechanisms remain unclear. Recent preclinical studies have focused on the fundamental mechanisms underlying the coexistence of chronic pain and cognitive decline. Pain chronification is accompanied by structural and functional changes in the neural substrate of cognition. Based on the developments in electrophysiology and optogenetics/chemogenetics, we summarized the relevant neural circuits involved in pain-induced cognitive impairment, as well as changes in connectivity and function in brain regions. We then present the cellular and molecular alternations related to pain-induced cognitive impairment in preclinical studies, mainly including modifications in neuronal excitability and structure, synaptic plasticity, glial cells and cytokines, neurotransmitters and other neurochemicals, and the gut-brain axis. Finally, we also discussed the potential treatment strategies and future research directions.
Collapse
Affiliation(s)
- Siyi Han
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei, China
| | - Jie Wang
- Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Wen Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei, China.
| | - Xuebi Tian
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei, China.
| |
Collapse
|
4
|
Pattison LA, Cloake A, Chakrabarti S, Hilton H, Rickman RH, Higham JP, Meng MY, Paine LW, Dannawi M, Qiu L, Ritoux A, Bulmer DC, Callejo G, Smith ESJ. Digging deeper into pain: an ethological behavior assay correlating well-being in mice with human pain experience. Pain 2024; 165:1761-1773. [PMID: 38452214 PMCID: PMC11247454 DOI: 10.1097/j.pain.0000000000003190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 03/09/2024]
Abstract
ABSTRACT The pressing need for safer, more efficacious analgesics is felt worldwide. Preclinical tests in animal models of painful conditions represent one of the earliest checkpoints novel therapeutics must negotiate before consideration for human use. Traditionally, the pain status of laboratory animals has been inferred from evoked nociceptive assays that measure their responses to noxious stimuli. The disconnect between how pain is tested in laboratory animals and how it is experienced by humans may in part explain the shortcomings of current pain medications and highlights a need for refinement. Here, we survey human patients with chronic pain who assert that everyday aspects of life, such as cleaning and leaving the house, are affected by their ongoing level of pain. Accordingly, we test the impact of painful conditions on an ethological behavior of mice, digging. Stable digging behavior was observed over time in naive mice of both sexes. By contrast, deficits in digging were seen after acute knee inflammation. The analgesia conferred by meloxicam and gabapentin was compared in the monosodium iodoacetate knee osteoarthritis model, with meloxicam more effectively ameliorating digging deficits, in line with human patients finding meloxicam more effective. Finally, in a visceral pain model, the decrease in digging behavior correlated with the extent of disease. Ultimately, we make a case for adopting ethological assays, such as digging, in studies of pain in laboratory animals, which we believe to be more representative of the human experience of pain and thus valuable in assessing clinical potential of novel analgesics in animals.
Collapse
Affiliation(s)
- Luke A. Pattison
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Alexander Cloake
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Sampurna Chakrabarti
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Helen Hilton
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Rebecca H. Rickman
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - James P. Higham
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Michelle Y. Meng
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Luke W. Paine
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Maya Dannawi
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Lanhui Qiu
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Anne Ritoux
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - David C. Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Gerard Callejo
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom. Dr. Chakrabarti is now with Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. Dr. Callejo is now with Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Parent HH, Niswender CM. Therapeutic Potential for Metabotropic Glutamate Receptor 7 Modulators in Cognitive Disorders. Mol Pharmacol 2024; 105:348-358. [PMID: 38423750 PMCID: PMC11026152 DOI: 10.1124/molpharm.124.000874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is the most highly conserved and abundantly expressed mGlu receptor in the human brain. The presynaptic localization of mGlu7, coupled with its low affinity for its endogenous agonist, glutamate, are features that contribute to the receptor's role in modulating neuronal excitation and inhibition patterns, including long-term potentiation, in various brain regions. These characteristics suggest that mGlu7 modulation may serve as a novel therapeutic strategy in disorders of cognitive dysfunction, including neurodevelopmental disorders that cause impairments in learning, memory, and attention. Primary mutations in the GRM7 gene have recently been identified as novel causes of neurodevelopmental disorders, and these patients exhibit profound intellectual and cognitive disability. Pharmacological tools, such as agonists, antagonists, and allosteric modulators, have been the mainstay for targeting mGlu7 in its endogenous homodimeric form to probe effects of its function and modulation in disease models. However, recent research has identified diversity in dimerization, as well as trans-synaptic interacting proteins, that also play a role in mGlu7 signaling and pharmacological properties. These novel findings represent exciting opportunities in the field of mGlu receptor drug discovery and highlight the importance of further understanding the functions of mGlu7 in complex neurologic conditions at both the molecular and physiologic levels. SIGNIFICANCE STATEMENT: Proper expression and function of mGlu7 is essential for learning, attention, and memory formation at the molecular level within neural circuits. The pharmacological targeting of mGlu7 is undergoing a paradigm shift by incorporating an understanding of receptor interaction with other cis- and trans- acting synaptic proteins, as well as various intracellular signaling pathways. Based upon these new findings, mGlu7's potential as a drug target in the treatment of cognitive disorders and learning impairments is primed for exploration.
Collapse
Affiliation(s)
- Harrison H Parent
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| | - Colleen M Niswender
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| |
Collapse
|
6
|
Mehsein Z, Kobaïter-Maarrawi S, Samaha H, El Shami M, Albeaini S, Maarrawi J. Right posterior insular epidural stimulation in rats with neuropathic pain induces a frequency-dependent and opioid system-mediated reduction of pain and its comorbid anxiety and depression. Prog Neuropsychopharmacol Biol Psychiatry 2024; 128:110845. [PMID: 37619765 DOI: 10.1016/j.pnpbp.2023.110845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/29/2023] [Accepted: 08/19/2023] [Indexed: 08/26/2023]
Abstract
Neuropathic pain (NP) is a sensory, emotional, and persistent disturbing experience caused by a lesion or disease of the somatosensory system which can lead when chronic to comorbidities such as anxiety and depression. Available treatments (pharmacotherapy, neurostimulation) have partial and unpredictable response; therefore, it seems necessary to find a new therapeutical approach that could alleviate most related symptoms and improve patients 'emotional state'. Posterior Insula seems to be a potential target of neurostimulation for pain relief. However, its effects on pain-related anxiety and depression remain unknown. Using rats with spared nerve injury (SNI), this study aims to elucidate the correlation between NP and anxio-depressive disorders, evaluate potential analgesic, anxiolytic, and antidepressant effects of right posterior insula stimulation (IS) using low (LF-IS, 50 Hz) or high (HF-IS, 150 Hz) frequency and assess endogenous opioid involvement in these effects. Results showed positive correlation between NP, anxiety, and depression. LF-IS reversed anhedonia and despair-like behavior through pain alleviation, whereas HF-IS only reduced anhedonia, all effects involving endogenous opioids. These findings support the link between NP and anxio-depressive disorders. Moreover, IS appears to have analgesic, anxiolytic and antidepressant effects mediated by the endogenous opioid system, making it a promising target for neurostimulation.
Collapse
Affiliation(s)
- Zeinab Mehsein
- Laboratory of Research in Neuroscience (LAREN), Pôle Technologie Santé (PTS), Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Sandra Kobaïter-Maarrawi
- Laboratory of Research in Neuroscience (LAREN), Pôle Technologie Santé (PTS), Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon.
| | - Hady Samaha
- Laboratory of Research in Neuroscience (LAREN), Pôle Technologie Santé (PTS), Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Mohamad El Shami
- Laboratory of Research in Neuroscience (LAREN), Pôle Technologie Santé (PTS), Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Sylvana Albeaini
- Laboratory of Research in Neuroscience (LAREN), Pôle Technologie Santé (PTS), Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Joseph Maarrawi
- Laboratory of Research in Neuroscience (LAREN), Pôle Technologie Santé (PTS), Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon; Department of Neurosurgery - Hôtel-Dieu de France Hospital, Beirut, Lebanon
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Design and Synthesis of New Quinazolin-4-one Derivatives with Negative mGlu 7 Receptor Modulation Activity and Antipsychotic-Like Properties. Int J Mol Sci 2023; 24:ijms24031981. [PMID: 36768302 PMCID: PMC9916658 DOI: 10.3390/ijms24031981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 01/21/2023] Open
Abstract
Following the glutamatergic theory of schizophrenia and based on our previous study regarding the antipsychotic-like activity of mGlu7 NAMs, we synthesized a new compound library containing 103 members, which were examined for NAM mGlu7 activity in the T-REx 293 cell line expressing a recombinant human mGlu7 receptor. Out of the twenty-two scaffolds examined, active compounds were found only within the quinazolinone chemotype. 2-(2-Chlorophenyl)-6-(2,3-dimethoxyphenyl)-3-methylquinazolin-4(3H)-one (A9-7, ALX-171, mGlu7 IC50 = 6.14 µM) was selective over other group III mGlu receptors (mGlu4 and mGlu8), exhibited satisfactory drug-like properties in preliminary DMPK profiling, and was further tested in animal models of antipsychotic-like activity, assessing the positive, negative, and cognitive symptoms. ALX-171 reversed DOI-induced head twitches and MK-801-induced disruptions of social interactions or cognition in the novel object recognition test and spatial delayed alternation test. On the other hand, the efficacy of the compound was not observed in the MK-801-induced hyperactivity test or prepulse inhibition. In summary, the observed antipsychotic activity profile of ALX-171 justifies the further development of the group of quinazolin-4-one derivatives in the search for a new drug candidate for schizophrenia treatment.
Collapse
|
10
|
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: 1.7] [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
|
11
|
Lombardo B, Pagani M, De Rosa A, Nunziato M, Migliarini S, Garofalo M, Terrile M, D’Argenio V, Galbusera A, Nuzzo T, Ranieri A, Vitale A, Leggiero E, Di Maio A, Barsotti N, Borello U, Napolitano F, Mandarino A, Carotenuto M, Heresco-Levy U, Pasqualetti M, Malatesta P, Gozzi A, Errico F, Salvatore F, Pastore L, Usiello A. D-aspartate oxidase gene duplication induces social recognition memory deficit in mice and intellectual disabilities in humans. Transl Psychiatry 2022; 12:305. [PMID: 35915065 PMCID: PMC9343392 DOI: 10.1038/s41398-022-02088-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/16/2022] [Accepted: 07/21/2022] [Indexed: 11/21/2022] Open
Abstract
The D-aspartate oxidase (DDO) gene encodes the enzyme responsible for the catabolism of D-aspartate, an atypical amino acid enriched in the mammalian brain and acting as an endogenous NMDA receptor agonist. Considering the key role of NMDA receptors in neurodevelopmental disorders, recent findings suggest a link between D-aspartate dysmetabolism and schizophrenia. To clarify the role of D-aspartate on brain development and functioning, we used a mouse model with constitutive Ddo overexpression and D-aspartate depletion. In these mice, we found reduced number of BrdU-positive dorsal pallium neurons during corticogenesis, and decreased cortical and striatal gray matter volume at adulthood. Brain abnormalities were associated with social recognition memory deficit at juvenile phase, suggesting that early D-aspartate occurrence influences neurodevelopmental related phenotypes. We corroborated this hypothesis by reporting the first clinical case of a young patient with severe intellectual disability, thought disorders and autism spectrum disorder symptomatology, harboring a duplication of a chromosome 6 region, including the entire DDO gene.
Collapse
Affiliation(s)
- Barbara Lombardo
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy ,grid.4691.a0000 0001 0790 385XDipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Marco Pagani
- grid.25786.3e0000 0004 1764 2907Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Arianna De Rosa
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Marcella Nunziato
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy ,grid.4691.a0000 0001 0790 385XDipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Sara Migliarini
- grid.5395.a0000 0004 1757 3729Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, 56126 Pisa, Italy
| | - Martina Garofalo
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy ,grid.9841.40000 0001 2200 8888Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Marta Terrile
- grid.5606.50000 0001 2151 3065Dipartimento di Oncologia, Biologia e Genetica, Università di Genova, 16132 Genoa, Italy ,grid.496862.70000 0004 0544 6263Present Address: Novartis Ireland ltd, D04A9N6 Dublin 4, Ireland
| | - Valeria D’Argenio
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy ,Dipartimento di Promozione delle Scienze Umane e della Qualità della Vita, Università San Raffaele, 00166 Rome, Italy
| | - Alberto Galbusera
- grid.25786.3e0000 0004 1764 2907Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Tommaso Nuzzo
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy ,grid.9841.40000 0001 2200 8888Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Annaluisa Ranieri
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy ,grid.4691.a0000 0001 0790 385XDipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Andrea Vitale
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy ,grid.4691.a0000 0001 0790 385XDipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Eleonora Leggiero
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Anna Di Maio
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Noemi Barsotti
- grid.5395.a0000 0004 1757 3729Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, 56126 Pisa, Italy
| | - Ugo Borello
- grid.5395.a0000 0004 1757 3729Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, 56126 Pisa, Italy
| | - Francesco Napolitano
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy ,grid.4691.a0000 0001 0790 385XDepartment of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy
| | - Alessandra Mandarino
- grid.9841.40000 0001 2200 8888Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80100 Naples, Italy
| | - Marco Carotenuto
- grid.9841.40000 0001 2200 8888Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80100 Naples, Italy
| | - Uriel Heresco-Levy
- grid.414060.70000 0004 0470 6676Research and Psychiatry Departments, Ezrath Nashim-Herzog Memorial Hospital, 9190501 Jerusalem, Israel ,grid.9619.70000 0004 1937 0538Hadassah Medical School, Hebrew University, 9190501 Jerusalem, Israel
| | - Massimo Pasqualetti
- grid.25786.3e0000 0004 1764 2907Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy ,grid.5395.a0000 0004 1757 3729Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, 56126 Pisa, Italy
| | - Paolo Malatesta
- grid.5606.50000 0001 2151 3065Dipartimento di Medicina Sperimentale, Università di Genova, 16132 Genoa, Italy ,grid.410345.70000 0004 1756 7871Ospedale Policlinico San Martino IRCCS, 16132 Genoa, Italy
| | - Alessandro Gozzi
- grid.25786.3e0000 0004 1764 2907Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Francesco Errico
- grid.4691.a0000 0001 0790 385XCEINGE Biotecnologie Avanzate, 80145 Naples, Italy ,grid.4691.a0000 0001 0790 385XDepartment of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy
| | - Francesco Salvatore
- CEINGE Biotecnologie Avanzate, 80145, Naples, Italy. .,Centro Interuniversitario per Malattie Multigeniche e Multifattoriali e loro modelli animali (Federico II, 80131, Naples; Tor Vergata, Rome and "G. D'Annunzio", Chieti-Pescara), Naples, Italy.
| | - Lucio Pastore
- CEINGE Biotecnologie Avanzate, 80145, Naples, Italy. .,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131, Naples, Italy.
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate, 80145, Naples, Italy. .,Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
12
|
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: 0.3] [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]
|
13
|
Zhang KL, Li SJ, Pu XY, Wu FF, Liu H, Wang RQ, Liu BZ, Li Z, Li KF, Qian NS, Yang YL, Yuan H, Wang YY. Targeted up-regulation of Drp1 in dorsal horn attenuates neuropathic pain hypersensitivity by increasing mitochondrial fission. Redox Biol 2021; 49:102216. [PMID: 34954498 PMCID: PMC8718665 DOI: 10.1016/j.redox.2021.102216] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/11/2021] [Accepted: 12/15/2021] [Indexed: 01/02/2023] Open
Abstract
Mitochondria play an essential role in pathophysiology of both inflammatory and neuropathic pain (NP), but the mechanisms are not yet clear. Dynamin-related protein 1 (Drp1) is broadly expressed in the central nervous system and plays a role in the induction of mitochondrial fission process. Spared nerve injury (SNI), due to the dysfunction of the neurons within the spinal dorsal horn (SDH), is the most common NP model. We explored the neuroprotective role of Drp1 within SDH in SNI. SNI mice showed pain behavior and anxiety-like behavior, which was associated with elevation of Drp1, as well as increased density of mitochondria in SDH. Ultrastructural analysis showed SNI induced damaged mitochondria into smaller perimeter and area, tending to be circular. Characteristics of vacuole in the mitochondria further showed SNI induced the increased number of vacuole, widened vac-perimeter and vac-area. Stable overexpression of Drp1 via AAV under the control of the Drp1 promoter by intraspinal injection (Drp1 OE) attenuated abnormal gait and alleviated pain hypersensitivity of SNI mice. Mitochondrial ultrastructure analysis showed that the increased density of mitochondria induced by SNI was recovered by Drp1 OE which, however, did not change mitochondrial morphology and vacuole parameters within SDH. Contrary to Drp1 OE, down-regulation of Drp1 in the SDH by AAV-Drp1 shRNA (Drp1 RNAi) did not alter painful behavior induced by SNI. Ultrastructural analysis showed the treatment by combination of SNI and Drp1 RNAi (SNI + Drp1 RNAi) amplified the damages of mitochondria with the decreased distribution density, increased perimeter and area, as well as larger circularity tending to be more circular. Vacuole data showed SNI + Drp1 RNAi increased vacuole density, perimeter and area within the SDH mitochondria. Our results illustrate that mitochondria within the SDH are sensitive to NP, and targeted mitochondrial Drp1 overexpression attenuates pain hypersensitivity. Drp1 offers a novel therapeutic target for pain treatment.
Collapse
Affiliation(s)
- Kun-Long Zhang
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China; Department of Rehabilitation Medicine, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Shu-Jiao Li
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xue-Yin Pu
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Fei-Fei Wu
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Liu
- Department of Human Anatomy, Yan-An University, Yan'an, 716000, China
| | - Rui-Qing Wang
- Department of Human Anatomy, Yan-An University, Yan'an, 716000, China
| | - Bo-Zhi Liu
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ze Li
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Kai-Feng Li
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China
| | - Nian-Song Qian
- Department of Oncology, First Medical Center, The General Hospital of the People's Liberation Army, Beijing, 100000, China
| | - Yan-Ling Yang
- Department of Liver and Gallbladder Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Hua Yuan
- Department of Rehabilitation Medicine, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Ya-Yun Wang
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China; State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
14
|
Targeting metabotropic glutamate receptors for the treatment of depression and other stress-related disorders. Neuropharmacology 2021; 196:108687. [PMID: 34175327 DOI: 10.1016/j.neuropharm.2021.108687] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022]
Abstract
The discovery of robust antidepressant effects of ketamine in refractory patients has led to increasing focus on agents targeting glutamatergic signaling as potential novel antidepressant strategy. Among the agents targeting the glutamatergic system, compounds acting at metabotropic glutamate (mGlu) receptors are among the most promising agents under studies for depressive disorders. Further, the receptor diversity, distinct distribution in the CNS, and ability to modulate the glutamatergic neurotransmission in the brain areas implicated in mood disorders make them an exciting target for stress-related disorders. In preclinical models, antidepressant and anxiolytic effects of mGlu5 negative allosteric modulators (NAMs) have been reported. Interestingly, mGlu2/3 receptor antagonists show fast and sustained antidepressant-like effects similar to that of ketamine in rodents. Excitingly, they can also induce antidepressant effects in the animal models of treatment-resistant depression and are devoid of the side-effects associated with ketamine. Unfortunately, clinical trials of both mGlu5 and mGlu2/3 receptor NAMs have been inconclusive, and additional trials using other compounds with suitable preclinical and clinical properties are needed. Although group III mGlu receptors have gained less attention, mGlu7 receptor ligands have been shown to induce antidepressant-like effects in rodents. Collectively, compounds targeting mGlu receptors provide an alternative approach to fill the outstanding clinical need for safer and more efficacious antidepressants. This article is part of the special Issue on "Glutamate Receptors - mGluRs".
Collapse
|
15
|
Lu CC, Nyam TTE, Kuo JR, Lee YL, Chio CC, Wang CC. The neuroprotective effects of AMN082 on neuronal apoptosis in rats after traumatic brain injury. BMC Neurosci 2021; 22:44. [PMID: 34171999 PMCID: PMC8228939 DOI: 10.1186/s12868-021-00649-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/10/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to investigate whether AMN082 exerts its neuroprotective effect by attenuating glutamate receptor-associated neuronal apoptosis and improving functional outcomes after traumatic brain injury (TBI). METHODS Anesthetized male Sprague-Dawley rats were divided into the sham-operated, TBI + vehicle, and TBI + AMN082 groups. AMN082 (10 mg/kg) was intraperitoneally injected 0, 24, or 48 h after TBI. In the 120 min after TBI, heart rate, mean arterial pressure, intracranial pressure (ICP), and cerebral perfusion pressure (CPP) were continuously measured. Motor function, the infarct volume, neuronal nitrosative stress-associated apoptosis, and N-methyl-D-aspartate receptor 2A (NR2A) and NR2B expression in the pericontusional cortex were measured on the 3rd day after TBI. RESULTS The results showed that the AMN082-treated group had a lower ICP and higher CPP after TBI. TBI-induced motor deficits, the increase in infarct volume, neuronal apoptosis, and 3-nitrotyrosine and inducible nitric oxide synthase expression in the pericontusional cortex were significantly improved by AMN082 therapy. Simultaneously, AMN082 increased NR2A and NR2B expression in neuronal cells. CONCLUSIONS We concluded that intraperitoneal injection of AMN082 for 3 days may ameliorate TBI by attenuating glutamate receptor-associated nitrosative stress and neuronal apoptosis in the pericontusional cortex. We suggest that AMN082 administration in the acute stage may be a promising strategy for TBI.
Collapse
Affiliation(s)
- Chung-Che Lu
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Tee-Tau Eric Nyam
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Jinn-Rung Kuo
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yao-Lin Lee
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Chung-Ching Chio
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Che-Chuan Wang
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
- Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| |
Collapse
|
16
|
Medeiros P, Dos Santos IR, Júnior IM, Palazzo E, da Silva JA, Machado HR, Ferreira SH, Maione S, Coimbra NC, de Freitas RL. An Adapted Chronic Constriction Injury of the Sciatic Nerve Produces Sensory, Affective, and Cognitive Impairments: A Peripheral Mononeuropathy Model for the Study of Comorbid Neuropsychiatric Disorders Associated with Neuropathic Pain in Rats. PAIN MEDICINE 2021; 22:338-351. [PMID: 32875331 DOI: 10.1093/pm/pnaa206] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Chronic constriction injury (CCI) is a model of neuropathic pain induced by four loose ligatures around the sciatic nerve. This work aimed to investigate the sensory, affective, cognitive, and motor changes induced by an adaptation of the CCI model by applying a single ligature around the sciatic nerve. METHODS Mechanical allodynia was measured from day 1 to day 28 postsurgery by the von Frey test. The beam walking test (BWT) was conducted weekly until 28 days after surgery. Anxiety- and depression-like behaviors, and cognitive performance were assessed through the open field (OF), forced swimming (FS), and novel object recognition (NOR) tests, respectively, 21 days after surgery. RESULTS The two CCI models, both Bennett and Xie's model (four ligatures of the sciatic nerve) and a modification of it (one ligature), induced mechanical allodynia, increased immobility in the FS, and reduced recognition index in the NOR. The exploratory behavior and time spent in the central part of the arena decreased, while the defensive behavior increased in the OF. The animals subjected to the two CCI models showed motor alterations in the BWT; however, autotomy was observed only in the group with four ligatures and not in the group with a single ligature. CONCLUSIONS Overall these results demonstrate that our adapted CCI model, using a single ligature around the sciatic nerve, induces sensory, affective, cognitive, and motor alterations comparable to the CCI model with four ligatures without generating autotomy. This adaptation to the CCI model may therefore represent an appropriate and more easily performed model for inducing neuropathic pain and study underlying mechanisms and effective treatments.
Collapse
Affiliation(s)
- Priscila Medeiros
- Laboratory of Neurosciences of Pain & Emotions and Neuroelectrophysiology Multi-User Centre, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ieda Regina Dos Santos
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Ivair Matias Júnior
- Laboratory of Pediatric Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Enza Palazzo
- Division of Pharmacology, Department of Experimental Medicine, University of Campania "L. Vanvitelli," Naples, Italy
| | - José Aparecido da Silva
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Psychology, Ribeirão Preto School of Philosophy, Sciences and Literature of the University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Psychology Department, Federal University of Juiz de Fora (UFJF-MG), Juiz de Fora, Minas Gerais, Brazil
| | - Hélio Rubens Machado
- Laboratory of Neurosciences of Pain & Emotions and Neuroelectrophysiology Multi-User Centre, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Laboratory of Pediatric Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sérgio Henrique Ferreira
- Laboratory of Neurosciences of Pain & Emotions and Neuroelectrophysiology Multi-User Centre, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Biomedical Sciences Institute, Federal University of Alfenas (UNIFAL-MG), Str. Gabriel Monteiro da Silva, Alfenas, Minas Gerais, Brazil
| | - Sabatino Maione
- Division of Pharmacology, Department of Experimental Medicine, University of Campania "L. Vanvitelli," Naples, Italy.,IRCCS Neuromed, 86077, Pozzilli-Caserta, Italy
| | - Norberto Cysne Coimbra
- Laboratory of Neurosciences of Pain & Emotions and Neuroelectrophysiology Multi-User Centre, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Renato Leonardo de Freitas
- Laboratory of Neurosciences of Pain & Emotions and Neuroelectrophysiology Multi-User Centre, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Psychology, Ribeirão Preto School of Philosophy, Sciences and Literature of the University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Biomedical Sciences Institute, Federal University of Alfenas (UNIFAL-MG), Str. Gabriel Monteiro da Silva, Alfenas, Minas Gerais, Brazil
| |
Collapse
|
17
|
Abstract
Supplemental Digital Content is Available in the Text. Analysis of multiple rodent RNAseq after nerve injury reveals a common gene signature, with suppression of endogenous opioid signalling and overlap with human pain genes The dorsal root ganglia (DRG) are key structures in nociception and chronic pain disorders. Several gene expression studies of DRG in preclinical pain models have been performed, but it is unclear if consistent gene changes are identifiable. We, therefore, compared several recent RNA-Seq data sets on the whole DRG in rodent models of nerve injury. Contrary to previous findings, we show hundreds of common differentially expressed genes and high positive correlation between studies, despite model and species differences. We also find, in contrast to previous studies, that 60% of the common rodent gene response after injury is likely to occur in nociceptors of the DRG. Substantial expression changes are observed at a 1-week time-point, with smaller changes in the same genes at a later 3- to 4-week time-point. However, a subset of genes shows a similar magnitude of changes at both early and late time-points, suggesting their potential involvement in the maintenance of chronic pain. These genes are centred around suppression of endogenous opioid signalling. Reversal of this suppression could allow endogenous and exogenous opioids to exert their analgesic functions and may be an important strategy for treating chronic pain disorders. Currently used drugs, such as amitriptyline and duloxetine, do not seem to appropriately modulate many of the critical pain genes and indeed may transcriptionally suppress endogenous opioid signalling further.
Collapse
|
18
|
Gan Z, Li H, Naser PV, Oswald MJ, Kuner R. Suppression of neuropathic pain and comorbidities by recurrent cycles of repetitive transcranial direct current motor cortex stimulation in mice. Sci Rep 2021; 11:9735. [PMID: 33958647 PMCID: PMC8102487 DOI: 10.1038/s41598-021-89122-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/12/2021] [Indexed: 11/16/2022] Open
Abstract
Transcranial, minimally-invasive stimulation of the primary motor cortex (M1) has recently emerged to show promise in treating clinically refractory neuropathic pain. However, there is a major need for improving efficacy, reducing variability and understanding mechanisms. Rodent models hold promise in helping to overcome these obstacles. However, there still remains a major divide between clinical and preclinical studies with respect to stimulation programs, analysis of pain as a multidimensional sensory-affective-motivational state and lack of focus on chronic phases of established pain. Here, we employed direct transcranial M1 stimulation (M1 tDCS) either as a single 5-day block or recurring blocks of repetitive stimulation over early or chronic phases of peripherally-induced neuropathic pain in mice. We report that repeated blocks of stimulation reverse established neuropathic mechanical allodynia more strongly than a single 5-day regime and also suppress cold allodynia, aversive behavior and anxiety without adversely affecting motor function over a long period. Activity mapping revealed highly selective alterations in the posterior insula, periaqueductal gray subdivisions and superficial spinal laminae in reversal of mechanical allodynia. Our preclinical data reveal multimodal analgesia and improvement in quality of life by multiple blocks of M1 tDCS and uncover underlying brain networks, thus helping promote clinical translation.
Collapse
Affiliation(s)
- Zheng Gan
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Han Li
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Paul Vincent Naser
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Manfred Josef Oswald
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Rohini Kuner
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
19
|
Moreno AM, Alemán F, Catroli GF, Hunt M, Hu M, Dailamy A, Pla A, Woller SA, Palmer N, Parekh U, McDonald D, Roberts AJ, Goodwill V, Dryden I, Hevner RF, Delay L, Gonçalves Dos Santos G, Yaksh TL, Mali P. Long-lasting analgesia via targeted in situ repression of Na V1.7 in mice. Sci Transl Med 2021; 13:eaay9056. [PMID: 33692134 PMCID: PMC8830379 DOI: 10.1126/scitranslmed.aay9056] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/14/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022]
Abstract
Current treatments for chronic pain rely largely on opioids despite their substantial side effects and risk of addiction. Genetic studies have identified in humans key targets pivotal to nociceptive processing. In particular, a hereditary loss-of-function mutation in NaV1.7, a sodium channel protein associated with signaling in nociceptive sensory afferents, leads to insensitivity to pain without other neurodevelopmental alterations. However, the high sequence and structural similarity between NaV subtypes has frustrated efforts to develop selective inhibitors. Here, we investigated targeted epigenetic repression of NaV1.7 in primary afferents via epigenome engineering approaches based on clustered regularly interspaced short palindromic repeats (CRISPR)-dCas9 and zinc finger proteins at the spinal level as a potential treatment for chronic pain. Toward this end, we first optimized the efficiency of NaV1.7 repression in vitro in Neuro2A cells and then, by the lumbar intrathecal route, delivered both epigenome engineering platforms via adeno-associated viruses (AAVs) to assess their effects in three mouse models of pain: carrageenan-induced inflammatory pain, paclitaxel-induced neuropathic pain, and BzATP-induced pain. Our results show effective repression of NaV1.7 in lumbar dorsal root ganglia, reduced thermal hyperalgesia in the inflammatory state, decreased tactile allodynia in the neuropathic state, and no changes in normal motor function in mice. We anticipate that this long-lasting analgesia via targeted in vivo epigenetic repression of NaV1.7 methodology we dub pain LATER, might have therapeutic potential in management of persistent pain states.
Collapse
Affiliation(s)
- Ana M Moreno
- Department of Bioengineering, University of California San Diego, San Diego, CA 92093, USA
| | - Fernando Alemán
- Department of Bioengineering, University of California San Diego, San Diego, CA 92093, USA
| | - Glaucilene F Catroli
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92093, USA
| | - Matthew Hunt
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92093, USA
| | - Michael Hu
- Department of Bioengineering, University of California San Diego, San Diego, CA 92093, USA
| | - Amir Dailamy
- Department of Bioengineering, University of California San Diego, San Diego, CA 92093, USA
| | - Andrew Pla
- Department of Bioengineering, University of California San Diego, San Diego, CA 92093, USA
| | - Sarah A Woller
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92093, USA
| | - Nathan Palmer
- Division of Biological Sciences, University of California San Diego , San Diego, CA 92093, USA
| | - Udit Parekh
- Department of Electrical Engineering, University of California San Diego , San Diego, CA 92093, USA
| | - Daniella McDonald
- Department of Bioengineering, University of California San Diego, San Diego, CA 92093, USA
- Biomedical Sciences Graduate Program, University of California San Diego, San Diego, San Diego, CA 92093, USA
| | - Amanda J Roberts
- Animal Models Core, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vanessa Goodwill
- Department of Neuropathology, University of California San Diego, San Diego, CA 92093, USA
| | - Ian Dryden
- Department of Neuropathology, University of California San Diego, San Diego, CA 92093, USA
| | - Robert F Hevner
- Department of Neuropathology, University of California San Diego, San Diego, CA 92093, USA
| | - Lauriane Delay
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92093, USA
| | | | - Tony L Yaksh
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92093, USA.
| | - Prashant Mali
- Department of Bioengineering, University of California San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
20
|
Kang JWM, Mor D, Keay KA. Nerve injury alters restraint-induced activation of the basolateral amygdala in male rats. Brain Struct Funct 2021; 226:1209-1227. [PMID: 33582845 DOI: 10.1007/s00429-021-02235-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 01/28/2021] [Indexed: 01/05/2023]
Abstract
The amygdala is critical for the production of appropriate responses towards emotional or stressful stimuli. It has a characteristic neuronal activation pattern to acute stressors. Chronic pain and acute stress have each been shown to independently modulate the activity of the amygdala. Few studies have investigated the effect of pain or injury, on amygdala activation to acute stress. This study investigated the effects of a neuropathic injury on the activation response of the amygdala to an acute restraint stress. Chronic constriction injury of the right sciatic nerve (CCI) was used to create neuropathic injury and a single brief 15-min acute restraint was used as an emotional/psychological stressor. All rats received cholera toxin B (CTB) retrograde tracer injections into the medial prefrontal cortex (mPFC) to assess if the amygdala to mPFC pathway was specifically regulated by the combination of neuropathic injury and acute stress. To assess differential patterns of activity in amygdala subregions, cFos expression was used as a marker for "acute", restraint triggered neuronal activation, and FosB/ΔFosB expression was used to reveal prolonged neuronal activation/sensitisation triggered by CCI. Restraint resulted in a characteristic increase in cFos expression in the medial amygdala, which was not altered by CCI. Rats with a CCI showed increased cFos expression in the basolateral amygdala (BLA), in response to an acute restraint stress, but not in neurons projecting to the prefrontal cortex. Further, CCI rats showed an increase in FosB/ΔFosB expression which was exclusive to the BLA. This increase likely reflects sensitisation of the BLA as a consequence of nerve injury which may contribute to heightened sensitivity of BLA neurons to acute emotional/ psychological stressors.
Collapse
Affiliation(s)
- James W M Kang
- Faculty of Medicine and Health, School of Medical Sciences (Neuroscience), The University of Sydney, Sydney, NSW, 2006, Australia. .,Brain and Mind Centre (M02G), 100 Mallet Street, Camperdown, NSW, 2050, Australia.
| | - David Mor
- Faculty of Medicine and Health, School of Medical Sciences (Neuroscience), The University of Sydney, Sydney, NSW, 2006, Australia
| | - Kevin A Keay
- Faculty of Medicine and Health, School of Medical Sciences (Neuroscience), The University of Sydney, Sydney, NSW, 2006, Australia.,Brain and Mind Centre (M02G), 100 Mallet Street, Camperdown, NSW, 2050, Australia
| |
Collapse
|
21
|
Boccella S, Guida F, Iannotta M, Iannotti FA, Infantino R, Ricciardi F, Cristiano C, Vitale RM, Amodeo P, Marabese I, Belardo C, de Novellis V, Paino S, Palazzo E, Calignano A, Di Marzo V, Maione S, Luongo L. 2-Pentadecyl-2-oxazoline ameliorates memory impairment and depression-like behaviour in neuropathic mice: possible role of adrenergic alpha2- and H3 histamine autoreceptors. Mol Brain 2021; 14:28. [PMID: 33557888 PMCID: PMC7871413 DOI: 10.1186/s13041-020-00724-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/28/2020] [Indexed: 12/20/2022] Open
Abstract
Neuropathic pain (NP) remains an untreatable disease due to the complex pathophysiology that involves the whole pain neuraxis including the forebrain. Sensory dysfunctions such as allodynia and hyperalgesia are only part of the symptoms associated with neuropathic pain that extend to memory and affectivity deficits. The development of multi-target molecules might be a promising therapeutic strategy against the symptoms associated with NP. 2-pentadecyl-2-oxazoline (PEA-OXA) is a plant-derived agent, which has shown effectiveness against chronic pain and associated neuropsychiatric disorders. The molecular mechanisms by which PEA-OXA exerts its effects are, however, only partially known. In the current study, we show that PEA-OXA, besides being an alpha2 adrenergic receptor antagonist, also acts as a modulator at histamine H3 receptors, and report data on its effects on sensory, affective and cognitive symptoms associated with the spared nerve injury (SNI) model of neuropathic pain in mice. Treatment for 14 days with PEA-OXA after the onset of the symptoms associated with neuropathic pain resulted in the following effects: (i) allodynia was decreased; (ii) affective/cognitive impairment associated with SNI (depression, spatial, and working memories) was counteracted; (iii) long-term potentiation in vivo in the lateral entorhinal cortex-dentate gyrus (perforant pathway, LPP) was ameliorated, (iv) hippocampal glutamate, GABA, histamine, norepinephrine and dopamine level alterations after peripheral nerve injury were reversed, (v) expression level of the TH positive neurons in the Locus Coeruleus were normalized. Thus, a 16-day treatment with PEA-OXA alleviates the sensory, emotional, cognitive, electrophysiological and neurochemical alterations associated with SNI-induced neuropathic pain.
Collapse
Affiliation(s)
- Serena Boccella
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Fabio Arturo Iannotti
- Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Flavia Ricciardi
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Claudia Cristiano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | | | - Pietro Amodeo
- Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Ida Marabese
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Carmela Belardo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Vito de Novellis
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Salvatore Paino
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Enza Palazzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Quebec City, Canada
| | - Sabatino Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
- IRCSS, Neuromed, Pozzilli, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138, Naples, Italy.
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy.
- IRCSS, Neuromed, Pozzilli, Italy.
| |
Collapse
|
22
|
Selective activation of metabotropic glutamate receptor 7 blocks paclitaxel-induced acute neuropathic pain and suppresses spinal glial reactivity in rats. Psychopharmacology (Berl) 2021; 238:107-119. [PMID: 33089875 DOI: 10.1007/s00213-020-05662-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 09/10/2020] [Indexed: 12/30/2022]
Abstract
RATIONALE Paclitaxel-induced acute pain syndrome (P-APS), characterized by deep muscle aches and arthralgia, occurs in more than 70% of patients who receive paclitaxel. P-APS can be debilitating for patients and lead to reductions and discontinuation of potentially curable therapy. Despite being relatively common in clinical practice, no clear treatment exists for P-APS and the underlying mechanisms remain poorly defined. Regulation of glutamatergic transmission by metabotropic glutamate receptors (mGluRs) has received growing attention with respect to its role in neuropathic pain. To our knowledge, no study has been conducted on alterations and functions of group III mGluR7 signaling in P-APS. OBJECTIVES In the present study, we determined whether a single administration of paclitaxel induces glutamatergic alterations and whether mGluR7 activation blocks paclitaxel-induced neuropathic pain by suppressing glial reactivity in the spinal cord. RESULTS A single paclitaxel injection dose-dependently induced acute mechanical and thermal hypersensitivity, and was associated with increased glutamate level accompanied by reduction in mGluR7 expression in the spinal cord. Selective activation of mGluR7 by its positive allosteric modulator, AMN082, blocked the development of paclitaxel-induced acute mechanical and thermal hypersensitivity, without affecting the normal pain behavior of control rats. Moreover, activation of mGluR7 by AMN082 inhibited glial reactivity and decreased pro-inflammatory cytokine release during P-APS. Abortion of spinal glial reaction to paclitaxel alleviated paclitaxel-induced acute mechanical and thermal hypersensitivity. CONCLUSIONS There results support the hypothesis that spinal mGluR7 signaling plays an important role in P-APS; Selective activation of mGluR7 by its positive allosteric modulator, AMN082, blocks P-APS in part by reducing spinal glial reactivity and neuroinflammatory process.
Collapse
|
23
|
Xiao C, Liu D, Du J, Guo Y, Deng Y, Hei Z, Li X. Early molecular alterations in anterior cingulate cortex and hippocampus in a rodent model of neuropathic pain. Brain Res Bull 2021; 166:82-91. [PMID: 33253785 DOI: 10.1016/j.brainresbull.2020.11.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 01/16/2023]
Abstract
Neuropathic pain is clinically associated with the development of mental disorders. However, the early molecular changes possibly related to the late-set depressive consequence of neuropathic pain were obscure so far. In this genome-wide study, we aimed to characterize the molecular mechanisms at the early and late stages of neuropathic pain. The genetic data from anterior cingulate cortex (ACC) tissues of neuropathic pain mice in Gene Expression Omnibus database were analyzed by weighted gene co-expression network analysis. Modules with clinical significance were respectively distinguished for mice at two and eight weeks after operation. The genes that co-expressed in modules from two postoperative time points were obtained, and annotated by gene ontology and pathway enrichment analyses. Moreover, the hub genes were identified from the protein-protein interaction network, and their expression levels were validated by molecular biology experiments. Overall, two modules were respectively found to be associated with the neuropathic pain mice with and without depressive consequence. A total of 20 genes co-expressed in both modules, and MAPK signaling pathway was the most significant pathway for these genes. Furtherly, Dusp1, c-Fos and Gadd45β were identified as the hub genes. At two weeks after sciatic nerve cuffing, Gadd45β was significantly downregulated at both mRNA and protein levels in ACC and hippocampus, while the significant upregulation was only observed in mRNA and protein levels for c-Fos in ACC. This study firstly compared the gene expression profiles between neuropathic pain animals with and without depressive-like behavior, and we suggested the early changes in the activities of MAPK signaling pathway, c-Fos and Gadd45β might be related to late-onset depressive behavior induced by peripheral nerve injury.
Collapse
Affiliation(s)
- Cuicui Xiao
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dezhao Liu
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jingyi Du
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yue Guo
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yifan Deng
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiang Li
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
24
|
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: 5.2] [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
|
25
|
Chronic pain impact on rodents’ behavioral repertoire. Neurosci Biobehav Rev 2020; 119:101-127. [DOI: 10.1016/j.neubiorev.2020.09.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/14/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022]
|
26
|
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: 4.4] [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
|
27
|
Kummer KK, Mitrić M, Kalpachidou T, Kress M. The Medial Prefrontal Cortex as a Central Hub for Mental Comorbidities Associated with Chronic Pain. Int J Mol Sci 2020; 21:E3440. [PMID: 32414089 PMCID: PMC7279227 DOI: 10.3390/ijms21103440] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic pain patients frequently develop and suffer from mental comorbidities such as depressive mood, impaired cognition, and other significant constraints of daily life, which can only insufficiently be overcome by medication. The emotional and cognitive components of pain are processed by the medial prefrontal cortex, which comprises the anterior cingulate cortex, the prelimbic, and the infralimbic cortex. All three subregions are significantly affected by chronic pain: magnetic resonance imaging has revealed gray matter loss in all these areas in chronic pain conditions. While the anterior cingulate cortex appears hyperactive, prelimbic, and infralimbic regions show reduced activity. The medial prefrontal cortex receives ascending, nociceptive input, but also exerts important top-down control of pain sensation: its projections are the main cortical input of the periaqueductal gray, which is part of the descending inhibitory pain control system at the spinal level. A multitude of neurotransmitter systems contributes to the fine-tuning of the local circuitry, of which cholinergic and GABAergic signaling are particularly emerging as relevant components of affective pain processing within the prefrontal cortex. Accordingly, factors such as distraction, positive mood, and anticipation of pain relief such as placebo can ameliorate pain by affecting mPFC function, making this cortical area a promising target region for medical as well as psychosocial interventions for pain therapy.
Collapse
Affiliation(s)
| | | | | | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.K.K.); (M.M.); (T.K.)
| |
Collapse
|
28
|
Guida F, De Gregorio D, Palazzo E, Ricciardi F, Boccella S, Belardo C, Iannotta M, Infantino R, Formato F, Marabese I, Luongo L, de Novellis V, Maione S. Behavioral, Biochemical and Electrophysiological Changes in Spared Nerve Injury Model of Neuropathic Pain. Int J Mol Sci 2020; 21:ijms21093396. [PMID: 32403385 PMCID: PMC7246983 DOI: 10.3390/ijms21093396] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 01/05/2023] Open
Abstract
Neuropathic pain is a pathological condition induced by a lesion or disease affecting the somatosensory system, with symptoms like allodynia and hyperalgesia. It has a multifaceted pathogenesis as it implicates several molecular signaling pathways involving peripheral and central nervous systems. Affective and cognitive dysfunctions have been reported as comorbidities of neuropathic pain states, supporting the notion that pain and mood disorders share some common pathogenetic mechanisms. The understanding of these pathophysiological mechanisms requires the development of animal models mimicking, as far as possible, clinical neuropathic pain symptoms. Among them, the Spared Nerve Injury (SNI) model has been largely characterized in terms of behavioral and functional alterations. This model is associated with changes in neuronal firing activity at spinal and supraspinal levels, and induces late neuropsychiatric disorders (such as anxious-like and depressive-like behaviors, and cognitive impairments) comparable to an advanced phase of neuropathy. The goal of this review is to summarize current findings in preclinical research, employing the SNI model as a tool for identifying pathophysiological mechanisms of neuropathic pain and testing pharmacological agent.
Collapse
Affiliation(s)
- Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
- Correspondence: (F.G.); (S.M.)
| | - Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montréal, QC H3A1A1, Canada;
| | - Enza Palazzo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Flavia Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Carmela Belardo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Monica Iannotta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Rosmara Infantino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Federica Formato
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Ida Marabese
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Livio Luongo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Vito de Novellis
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
- Correspondence: (F.G.); (S.M.)
| |
Collapse
|
29
|
PCC0208009, an indirect IDO1 inhibitor, alleviates neuropathic pain and co-morbidities by regulating synaptic plasticity of ACC and amygdala. Biochem Pharmacol 2020; 177:113926. [PMID: 32217098 DOI: 10.1016/j.bcp.2020.113926] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Indoleamine 2, 3-dioxygenase 1 (IDO1) has been linked to neuropathic pain and IDO1 inhibitors have been shown to reduce pain in animals. Some studies have indicated that IDO1 expression increased after neuropathic pain in hippocampus and spinal cord, whether these changes existing in anterior cingulate cortex (ACC) and amygdala remains obscure and how IDO1 inhibition leads to analgesia is largely unknown. Here, we evaluated the antinociceptive effect of PCC0208009, an indirect IDO1 inhibitor, on neuropathic pain and examined the related neurobiological mechanisms. EXPERIMENTAL APPROACH The effects of PCC0208009 on pain, cognition and anxiogenic behaviors were evaluated in a rat model of neuropathic pain. Motor disorder, sedation and somnolence were also assessed. Biochemical techniques were used to measure IDO1-mediated signaling changes in ACC and amygdala. KEY RESULTS In rats receiving spinal nerve ligation (SNL), IDO1 expression level was increased in ACC and amygdala. PCC0208009 attenuated pain-related behaviors in the formalin test and SNL model and increased cognition and anxiogenic behaviors in SNL rats at doses that did not affect locomotor activity and sleeping. PCC0208009 inhibited IDO1 expression in ACC and amygdala by inhibiting the IL-6-JAK2/STAT3-IDO1-GCN2-IL-6 pathway. In addition, PCC0208009 reversed synaptic plasticity at the functional and structural levels by suppressing NMDA2B receptor and CDK5/MAP2 or CDK5/Tau pathway in ACC and amygdala. CONCLUSION AND IMPLICATIONS These results support the role of IDO1-mediated molecular mechanisms in neuropathic pain and suggest that the IDO1 inhibitor PCC0208009 demonstrates selective pain suppression and could be a useful pharmacological therapy for neuropathic pain.
Collapse
|
30
|
Won S, Park K, Lim H, Lee SJ. Sexual dimorphism in cognitive disorders in a murine model of neuropathic pain. Behav Brain Funct 2020; 16:1. [PMID: 31901234 PMCID: PMC6942364 DOI: 10.1186/s12993-019-0164-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/18/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A sex-difference in susceptibility to chronic pain is well-known. Although recent studies have begun to reveal the sex-dependent mechanisms of nerve injury-induced pain sensitization, sex differences in the affective and cognitive brain dysfunctions associated with chronic pain have not been investigated. Therefore, we tested whether chronic pain leads to affective and cognitive disorders in a mouse neuropathic pain model and whether those disorders are sexually dimorphic. METHODS Chronic neuropathic pain was induced in male and female mice by L5 spinal nerve transection (SNT) injury. Pain sensitivity was measured with the von Frey test. Affective behaviors such as depression and anxiety were assessed by the forced swim, tail suspension, and open field tests. Cognitive brain function was assessed with the Morris water maze and the novel object location and novel object recognition tests. RESULTS Mechanical allodynia was induced and maintained for up to 8 weeks after SNT in both male and female mice. Depressive- and anxiety-like behaviors were observed 8 weeks post-SNT injury regardless of sex. Chronic pain-induced cognitive deficits measured with the Morris water maze and novel object location test were seen only in male mice, not in female mice. CONCLUSIONS Chronic neuropathic pain is accompanied by anxiety- and depressive-like behaviors in a mouse model regardless of sex, and male mice are more vulnerable than female mice to chronic pain-associated cognitive deficits.
Collapse
Affiliation(s)
- Soonmi Won
- Department of Neuroscience and Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Keebum Park
- Department of Neuroscience and Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea.,Interdisciplinary Program in Neuroscience, College of Natural Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyoungsub Lim
- Department of Neuroscience and Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Sung Joong Lee
- Department of Neuroscience and Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea. .,Interdisciplinary Program in Neuroscience, College of Natural Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
31
|
The gap junction inhibitor INI-0602 attenuates mechanical allodynia and depression-like behaviors induced by spared nerve injury in rats. Neuroreport 2019; 30:369-377. [PMID: 30741784 DOI: 10.1097/wnr.0000000000001209] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gap junctions (GJs) are novel molecular targets for pain therapeutics due to their pain-promoting function. INI-0602, a new GJ inhibitor, exerts a neuroprotective role, while its role in neuropathic pain is unclear. The objective was to investigate the analgesic role and mechanisms of INI-0602 in neuropathic pain induced by spared nerve injury (SNI), and whether INI-0602 attenuated pain-induced depression-like behaviors. Rats were randomly assigned to saline treatment groups (sham+NS and SNI+NS) or INI-0602 treatment groups (sham+INI-0602 and SNI+INI-0602). The von Frey test was used to assess pain behavior, and the sucrose preference test, the forced swimming test, and the tail suspension test were used to assess depression-like behaviors. Gap junction intercellular communication (GJIC) was measured by parachute assay. Western blots were used to determine the protein expression. In vitro, INI-0602 significantly suppressed GJIC by decreasing connexin43 and connexin32 expression. In vivo, INI-0602 significantly suppressed mechanical allodynia during initiation (7 days after SNI) and the maintenance phase (21 days after SNI) and simultaneously attenuated accompanying depression-like behaviors. Furthermore, INI-0602 markedly suppressed the activation of astrocytes and microglia on days 7 and 21 by reducing GJIC. Finally, INI-0602 reversed the changes in the brain-derived neurotrophic factor and Nr2b subunits of the N-methyl-D-aspartate receptor in SNI rats, suggesting that these effects of INI-0602 were related to its analgesic effect. Our findings demonstrated that blocking GJs with INI-0602 attenuated mechanical pain hypersensitivity and related depression-like behaviors in SNI rats by reducing glial activation.
Collapse
|
32
|
N-methyl-d-aspartate Receptors in the Prelimbic Cortex are Critical for the Maintenance of Neuropathic Pain. Neurochem Res 2019; 44:2068-2080. [DOI: 10.1007/s11064-019-02843-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 06/27/2019] [Accepted: 07/04/2019] [Indexed: 12/13/2022]
|
33
|
Belardo C, Iannotta M, Boccella S, Rubino RC, Ricciardi F, Infantino R, Pieretti G, Stella L, Paino S, Marabese I, Maisto R, Luongo L, Maione S, Guida F. Oral Cannabidiol Prevents Allodynia and Neurological Dysfunctions in a Mouse Model of Mild Traumatic Brain Injury. Front Pharmacol 2019; 10:352. [PMID: 31040777 PMCID: PMC6476923 DOI: 10.3389/fphar.2019.00352] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/21/2019] [Indexed: 01/28/2023] Open
Abstract
Neurological dysfunctions are the most impactful and persistent consequences of traumatic brain injury (TBI). Indeed, previous reports suggest that an association between TBI and chronic pain syndromes, as well anxio-depressive behaviors, tends to be more common in patients with mild forms of TBI. At present, no effective treatment options are available for these symptoms. In the present study, we used a weight drop mild TBI mouse model to investigate the effect of a commercially available 10% Cannabidiol (CBD) oil on both the sensorial and neuropsychiatric dysfunctions associated with mild TBI through behavioral and biomolecular approaches. TBI mice developed chronic pain associated with anxious and aggressive behavior, followed by a late depressive-like behavior and impaired social interaction. Such behaviors were related with specific changes in neurotransmitters release at cortical levels. CBD oral treatment restored the behavioral alterations and partially normalized the cortical biochemical changes. In conclusion, our data show some of the brain modifications probably responsible for the behavioral phenotype associated with TBI and suggest the CBD as a pharmacological tool to improve neurological dysfunctions caused by the trauma.
Collapse
Affiliation(s)
- Carmela Belardo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Flavia Ricciardi
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gorizio Pieretti
- Department of Plastic Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Salvatore Paino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosa Maisto
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
34
|
Abstract
A great need exists for the identification of new effective analgesics to treat sustained pain. However, most preclinical nociceptive assays measure behavioral responses evoked by noxious stimuli (ie, pain-stimulated behavior), which presents a challenge to distinguish between motor impairing and antinociceptive effects of drugs. Here, we demonstrate that chronic constriction injury (CCI) of the sciatic nerve elicits common pain-stimulated responses (ie, mechanical allodynia and thermal hyperalgesia) as well as reduces marble burying/digging behaviors that occur during the early stages of the neuropathy and resolve within 1 week. Although drugs representing distinct classes of analgesics (ie, morphine, valdecoxib, and gabapentin) reversed both CCI-induced and CCI-depressed nociceptive measures, diazepam lacked antinociceptive effects in all assays and the kappa-opioid receptor agonist U69593 reversed pain-stimulated, but not pain-depressed behaviors. In addition, we tested drugs targeting distinct components of the endocannabinoid system, including agonists at cannabinoid receptors type 1 (CB1) and type 2 (CB2), as well as inhibitors of the endocannabinoid-regulating enzymes fatty acid amide hydrolase and monoacylglycerol lipase. Each of these drugs reversed all CCI-induced nociceptive measures, with the exception of the fatty acid amide hydrolase inhibitor that reversed pain-stimulated behaviors, only. These findings support the use of the mouse marble-burying assay as a model of pain-depressed behavior within the first week of sciatic nerve injury to examine candidate analgesics. These data also support existing preclinical research that cannabinoid receptor agonists and inhibitors of endocannabinoid-regulating enzymes merit consideration for the treatment of pain.
Collapse
|
35
|
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: 26.3] [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
|
36
|
Vázquez-Villa H, Trabanco AA. Progress toward allosteric ligands of metabotropic glutamate 7 (mGlu7) receptor: 2008-present. MEDCHEMCOMM 2019; 10:193-199. [PMID: 30881607 PMCID: PMC6390470 DOI: 10.1039/c8md00524a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/11/2018] [Indexed: 01/01/2023]
Abstract
Metabotropic glutamate type 7 (mGlu7) receptor is a member of the group III family of mGlu receptors. It is widely distributed in the central nervous system (CNS) and is preferentially expressed on presynaptic nerve terminals where it is thought to play a critical role in modulating normal neuronal function and synaptic transmission, making it particularly relevant in neuropharmacology. The lack of small-molecule mGlu7 ligands with adequate potency, selectivity and drug-like properties has resulted in difficulties in the preclinical validation of mGlu7 modulation in disease models. In the last decade, allosteric modulators of mGlu7 receptors have emerged as valuable tools with good potency, selectivity and physicochemical properties to study and unleash the therapeutic potential of mGlu7 receptors. This review focusses on the medicinal chemistry of mGlu7 receptor allosteric ligands discovered since 2008.
Collapse
Affiliation(s)
- Henar Vázquez-Villa
- Departamento de Química Orgánica , Facultad de Ciencias Químicas , Universidad Complutense de Madrid , E-28040 Madrid , Spain .
| | - Andrés A Trabanco
- Discovery Sciences , Medicinal Chemistry Department , Janssen Research & Development , c/ Jarama 75A , 45007 Toledo , Spain .
| |
Collapse
|
37
|
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: 11.5] [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
|
38
|
Belardo C, Iannotta M, Boccella S, Rubino RC, Ricciardi F, Infantino R, Pieretti G, Stella L, Paino S, Marabese I, Maisto R, Luongo L, Maione S, Guida F. Oral Cannabidiol Prevents Allodynia and Neurological Dysfunctions in a Mouse Model of Mild Traumatic Brain Injury. Front Pharmacol 2019. [PMID: 31040777 DOI: 10.3389/fphar.2019.00352/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Neurological dysfunctions are the most impactful and persistent consequences of traumatic brain injury (TBI). Indeed, previous reports suggest that an association between TBI and chronic pain syndromes, as well anxio-depressive behaviors, tends to be more common in patients with mild forms of TBI. At present, no effective treatment options are available for these symptoms. In the present study, we used a weight drop mild TBI mouse model to investigate the effect of a commercially available 10% Cannabidiol (CBD) oil on both the sensorial and neuropsychiatric dysfunctions associated with mild TBI through behavioral and biomolecular approaches. TBI mice developed chronic pain associated with anxious and aggressive behavior, followed by a late depressive-like behavior and impaired social interaction. Such behaviors were related with specific changes in neurotransmitters release at cortical levels. CBD oral treatment restored the behavioral alterations and partially normalized the cortical biochemical changes. In conclusion, our data show some of the brain modifications probably responsible for the behavioral phenotype associated with TBI and suggest the CBD as a pharmacological tool to improve neurological dysfunctions caused by the trauma.
Collapse
Affiliation(s)
- Carmela Belardo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Flavia Ricciardi
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gorizio Pieretti
- Department of Plastic Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Salvatore Paino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosa Maisto
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
39
|
Fisher NM, Seto M, Lindsley CW, Niswender CM. Metabotropic Glutamate Receptor 7: A New Therapeutic Target in Neurodevelopmental Disorders. Front Mol Neurosci 2018; 11:387. [PMID: 30405350 PMCID: PMC6206046 DOI: 10.3389/fnmol.2018.00387] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/01/2018] [Indexed: 12/27/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are characterized by a wide range of symptoms including delayed speech, intellectual disability, motor dysfunction, social deficits, breathing problems, structural abnormalities, and epilepsy. Unfortunately, current treatment strategies are limited and innovative new approaches are sorely needed to address these complex diseases. The metabotropic glutamate receptors are a class of G protein-coupled receptors that act to modulate neurotransmission across many brain structures. They have shown great promise as drug targets for numerous neurological and psychiatric diseases. Moreover, the development of subtype-selective allosteric modulators has allowed detailed studies of each receptor subtype. Here, we focus on the metabotropic glutamate receptor 7 (mGlu7) as a potential therapeutic target for NDDs. mGlu7 is expressed widely throughout the brain in regions that correspond to the symptom domains listed above and has established roles in synaptic physiology and behavior. Single nucleotide polymorphisms and mutations in the GRM7 gene have been associated with idiopathic autism and other NDDs in patients. In rodent models, existing literature suggests that decreased mGlu7 expression and/or function may lead to symptoms that overlap with those of NDDs. Furthermore, potentiation of mGlu7 activity has shown efficacy in a mouse model of Rett syndrome. In this review, we summarize current findings that provide rationale for the continued development of mGlu7 modulators as potential therapeutics.
Collapse
Affiliation(s)
- Nicole M Fisher
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Mabel Seto
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Craig W Lindsley
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.,Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Colleen M Niswender
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
40
|
Boccella S, Cristiano C, Romano R, Iannotta M, Belardo C, Farina A, Guida F, Piscitelli F, Palazzo E, Mazzitelli M, Imperatore R, Tunisi L, de Novellis V, Cristino L, Di Marzo V, Calignano A, Maione S, Luongo L. Ultra-micronized palmitoylethanolamide rescues the cognitive decline-associated loss of neural plasticity in the neuropathic mouse entorhinal cortex-dentate gyrus pathway. Neurobiol Dis 2018; 121:106-119. [PMID: 30266286 DOI: 10.1016/j.nbd.2018.09.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/10/2018] [Accepted: 09/24/2018] [Indexed: 02/08/2023] Open
Abstract
Chronic pain is associated with cognitive deficits. Palmitoylethanolamide (PEA) has been shown to ameliorate pain and pain-related cognitive impairments by restoring glutamatergic synapses functioning in the spared nerve injury (SNI) of the sciatic nerve in mice. SNI reduced mechanical and thermal threshold, spatial memory and LTP at the lateral entorhinal cortex (LEC)-dentate gyrus (DG) pathway. It decreased also postsynaptic density, volume and dendrite arborization of DG and increased the expression of metabotropic glutamate receptor 1 and 7 (mGluR1 and mGluR7), of the GluR1, GluR1s845 and GluR1s831 subunits of AMPA receptor and the levels of glutamate in the DG. The level of the endocannabinoid 2-arachidonoylglycerol (2-AG) was instead increased in the LEC. Chronic treatment with PEA, starting from when neuropathic pain was fully developed, was able to reverse mechanical allodynia and thermal hyperalgesia, memory deficit and LTP in SNI wild type, but not in PPARα null, mice. PEA also restored the level of glutamate and the expression of phosphorylated GluR1 subunits, postsynaptic density and neurogenesis. Altogether, these results suggest that neuropathic pain negatively affects cognitive behavior and related LTP, glutamatergic synapse and synaptogenesis in the DG. In these conditions PEA treatment alleviates pain and cognitive impairment by restoring LTP and synaptic maladaptative changes in the LEC-DG pathway. These outcomes open new perspectives for the use of the N-acylethanolamines, such as PEA, for the treatment of neuropathic pain and its central behavioural sequelae.
Collapse
Affiliation(s)
- Serena Boccella
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Claudia Cristiano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Rosaria Romano
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Carmela Belardo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Antonio Farina
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Enza Palazzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Roberta Imperatore
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Lea Tunisi
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Vito de Novellis
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy.
| |
Collapse
|
41
|
Cieślik P, Woźniak M, Kaczorowska K, Brański P, Burnat G, Chocyk A, Bobula B, Gruca P, Litwa E, Pałucha-Poniewiera A, Wąsik A, Pilc A, Wierońska J. Negative Allosteric Modulators of mGlu 7 Receptor as Putative Antipsychotic Drugs. Front Mol Neurosci 2018; 11:316. [PMID: 30294258 PMCID: PMC6158327 DOI: 10.3389/fnmol.2018.00316] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/16/2018] [Indexed: 01/08/2023] Open
Abstract
The data concerning antipsychotic-like activity of negative allosteric modulators (NAMs)/antagonists of mGlu7 receptors are limited. The only available ligands for this receptor are MMPIP and ADX71743. In the present studies, we used stable cell line expressing mGlu7 receptor and it was shown that both compounds dose-dependently potentiated forskolin elevated cAMP concentration in the T-REx 293 cells, showing their inverse agonist properties. Subsequently, pharmacokinetic studies were performed. Both compounds were given intraperitoneally (i.p.) at the dose of 10 mg/kg and reached Cmax 0.25-0.5 h after administration, and then they declined rapidly, ADX71743 being almost undetectable 2 h after administration, while the concentration of MMPIP was still observed, suggesting that the concentration of MMPIP was more stable. Finally, we investigated the role of both mGlu7 receptor NAMs in animal models of schizophrenia. Behavioral tests commonly used in antipsychotic drug discovery were conducted. Both tested compounds dose-dependently inhibited MK-801-induced hyperactivity (MMPIP at 15 mg/kg; ADX at 5 and 15 mg/kg) and DOI-induced head twitches (MMPIP at 5, 10, 15 mg/kg; ADX at 2.5, 5, 10 mg/kg). Moreover, the same effects were noticed in novel object recognition test, where MMPIP (5, 10, 15 mg/kg) and ADX71743 (1, 5, 15 mg/kg) reversed MK-801-induced disturbances. In the social interaction test, antipsychotic activity was observed only for ADX71743 (5, 15 mg/kg). ADX71743 at the dose 2.5 mg/kg reversed MK-801-induced disruption in prepulse inhibition while MMPIP at 10 mg/kg reversed MK-801-induced disruption in spatial delayed alternation. The present studies showed that mGlu7 receptor may be considered as a putative target for antipsychotic drugs, though more studies are needed due to limited number of available ligands.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Joanna Wierońska
- Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
42
|
Kiritoshi T, Neugebauer V. Pathway-Specific Alterations of Cortico-Amygdala Transmission in an Arthritis Pain Model. ACS Chem Neurosci 2018; 9:2252-2261. [PMID: 29630339 PMCID: PMC6146017 DOI: 10.1021/acschemneuro.8b00022] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Medial prefrontal cortex (mPFC) and amygdala are closely interconnected brain areas that play a key role in cognitive-affective aspects of pain through their reciprocal interactions. Clinical and preclinical evidence suggests that dysfunctions in the mPFC-amygdala circuitry underlie pain-related cognitive-affective deficits. However, synaptic mechanisms of pain-related changes in these long-range pathways are largely unknown. Here we used optogenetics and brain slice physiology to analyze synaptic transmission in different types of amygdala neurons driven by inputs from infralimbic (IL) and prelimbic (PL) subdivisions of the mPFC. We found that IL inputs evoked stronger synaptic inhibition of neurons in the latero-capsular division of the central nucleus (CeLC) of the amygdala than PL inputs, and this inhibition was impaired in an arthritis pain model. Furthermore, inhibition-excitation ratio in basolateral amygdala neurons was increased in the pain model in the IL pathway but not in the PL pathway. These results suggest that IL rather than PL controls CeLC activity, and that changes in this acute pain model occur predominantly in the IL-amygdala pathway.
Collapse
Affiliation(s)
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience
- Center of Excellence for Translational Neuroscience and Therapeutics Texas Tech University Health Sciences Center (TTUHSC), School of Medicine 3601 4th Street, Lubbock, TX 79430-6592
| |
Collapse
|
43
|
Standard analgesics reverse burrowing deficits in a rat CCI model of neuropathic pain, but not in models of type 1 and type 2 diabetes-induced neuropathic pain. Behav Brain Res 2018; 350:129-138. [DOI: 10.1016/j.bbr.2018.04.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/18/2018] [Accepted: 04/27/2018] [Indexed: 11/18/2022]
|
44
|
Jantas D, Lech T, Gołda S, Pilc A, Lasoń W. New evidences for a role of mGluR7 in astrocyte survival: Possible implications for neuroprotection. Neuropharmacology 2018; 141:223-237. [PMID: 30170084 DOI: 10.1016/j.neuropharm.2018.08.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/14/2018] [Accepted: 08/26/2018] [Indexed: 01/18/2023]
Abstract
A specific activation of metabotropic glutamate receptor 7 (mGluR7) has been shown to be neuroprotective in various models of neuronal cell damage, however, its role in glia cell survival has not been studied, yet. Thus, we performed comparative experiments estimating protective effects of the mGluR7 allosteric agonist AMN082 in glia, neuronal and neuronal-glia cell cultures against various harmful stimuli. First, the transcript levels of mGluR7 and other subtypes of group II and III mGluRs in cortical neuronal, neuronal-glia and glia cell cultures have been measured by qPCR method. Next, we demonstrated that AMN082 with similar efficiency attenuated the glia cell damage evoked by staurosporine (St) and doxorubicin (Dox). The AMN082-mediated glioprotection was mGluR7-dependent and associated with decreased DNA fragmentation without involvement of caspase-3 inhibition. Moreover, the inhibitors of PI3K/Akt and MAPK/ERK1/2 pathways blocked the protective effect of AMN082. In neuronal and neuronal-glia cell cultures in the model of glutamate (Glu)- but not St-evoked cell damage, we showed a significant glia contribution to mGluR7-mediated neuroprotection. Finally, by using glia and neuronal cells derived from mGluR7+/+ and mGluR7-/- mice we demonstrated a higher cell-damaging effect of St and Dox in mGluR7-deficient glia but not in neurons (cerebellar granule cells). Our present data showed for the first time a glioprotective potential of AMN082 underlain by mechanisms involving the activation of PI3K/Akt and MAPK/ERK1/2 pathways and pro-survival role of mGluR7 in glia cells. These findings together with the confirmed neuroprotective properties of AMN082 justify further research on mGluR7-targeted therapies for various CNS disorders.
Collapse
Affiliation(s)
- Danuta Jantas
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, PL, 31-343, Kraków, Poland.
| | - Tomasz Lech
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, PL, 31-343, Kraków, Poland
| | - Sławomir Gołda
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, PL, 31-343, Kraków, Poland
| | - Andrzej Pilc
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, PL, 31-343, Kraków, Poland
| | - Władysław Lasoń
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, PL, 31-343, Kraków, Poland
| |
Collapse
|
45
|
Metabotropic glutamate receptor subtype 7 in the dorsal striatum oppositely modulates pain in sham and neuropathic rats. Neuropharmacology 2018; 135:86-99. [PMID: 29505788 DOI: 10.1016/j.neuropharm.2018.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 02/23/2018] [Accepted: 03/01/2018] [Indexed: 11/24/2022]
Abstract
The study investigated the role of the metabotropic glutamate receptor subtype 7 (mGluR7) in pain signalling in the dorsal striatum of sham and neuropathic rats. Supraspinal circuitries involved in the dorsal striatum control of pain were also explored. In the sham rats, microinjection of N,N'-bis(diphenylmethyl)-1,2-ethanediamine (AMN082), a selective mGluR7 positive allosteric modulator, into the dorsal striatum, facilitated pain, increased the activity of the ON cells and inhibited the activity of the OFF cells in the rostral ventromedial medulla, and decreased glutamate levels in the dorsal striatum. Conversely, AMN082 inhibited pain and the activity of the ON cells while increased the activity of the OFF cells in rats with spared nerve injury (SNI) of the sciatic nerve. AMN082 also decreased glutamate levels in the dorsal striatum of SNI rats. The effect of AMN082 on mechanical allodynia and glutamate release was blocked by 6-(2,4-dimethylphenyl)-2-ethyl-6,7-dihydro-4(5H)-benzoxazolone (ADX71743), a selective mGluR7 negative allosteric modulator. Moreover, in the sham rats, AMN082 increased the activity of total nociceptive convergent neurons in the dorsal reticular nucleus while in the SNI rats, such activity was decreased. The administration of lidocaine into the subthalamic nucleus abolished the effect of AMN082 on the total nociceptive convergent neurons in the sham rats but not in the SNI rats. Thus, the dual effect of mGluR7 in facilitating or inhibiting pain responses may be due to the recruitment of different pathways of the basal ganglia, the indirect or direct pathway, in physiological or pathological conditions, respectively.
Collapse
|
46
|
Abstract
This chapter describes surgical procedures for the induction of neuropathic pain using an animal model (rat or mouse) of spared nerve injury. In addition to technical details of the surgical technique, details of anesthesia and perioperative care are also included.
Collapse
|
47
|
In vivo evaluation of the hippocampal glutamate, GABA and the BDNF levels associated with spatial memory performance in a rodent model of neuropathic pain. Physiol Behav 2017; 175:97-103. [DOI: 10.1016/j.physbeh.2017.03.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/18/2017] [Accepted: 03/18/2017] [Indexed: 11/22/2022]
|
48
|
Guo B, Wang J, Yao H, Ren K, Chen J, Yang J, Cai G, Liu H, Fan Y, Wang W, Wu S. Chronic Inflammatory Pain Impairs mGluR5-Mediated Depolarization-Induced Suppression of Excitation in the Anterior Cingulate Cortex. Cereb Cortex 2017; 28:2118-2130. [DOI: 10.1093/cercor/bhx117] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Baolin Guo
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Jiaqi Wang
- Cadet Brigade, Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Han Yao
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Keke Ren
- School of life Sciences, Yan’an University, Yan’an 716000, P.R. China
| | - Jing Chen
- Department of Anatomy and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Jing Yang
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Guohong Cai
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Haiying Liu
- Cadet Brigade, Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Yunlong Fan
- Cadet Brigade, Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Wenting Wang
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, P.R. China
| | - Shengxi Wu
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, P.R. China
| |
Collapse
|
49
|
Palazzo E, Marabese I, Luongo L, Guida F, de Novellis V, Maione S. Nociception modulation by supraspinal group III metabotropic glutamate receptors. J Neurochem 2017; 141:507-519. [DOI: 10.1111/jnc.13725] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 02/02/2023]
Affiliation(s)
- Enza Palazzo
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Ida Marabese
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Livio Luongo
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Francesca Guida
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Vito de Novellis
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Sabatino Maione
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| |
Collapse
|
50
|
Hellyer S, Leach K, Gregory KJ. Neurobiological insights and novel therapeutic opportunities for CNS disorders from mGlu receptor allosteric and biased modulation. Curr Opin Pharmacol 2017; 32:49-55. [DOI: 10.1016/j.coph.2016.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 11/30/2022]
|