1
|
Ma L, Sun D, Wen S, Yuan J, Li J, Tan X, Cao S. PSD-95 Protein: A Promising Therapeutic Target in Chronic Pain. Mol Neurobiol 2025; 62:3361-3375. [PMID: 39285025 DOI: 10.1007/s12035-024-04485-x] [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: 10/04/2023] [Accepted: 09/04/2024] [Indexed: 02/04/2025]
Abstract
Chronic pain, as a social public health problem, has a serious impact on the quality of patients' life. Currently, the main drugs used to treat chronic pain are opioids, antipyretic, and nonsteroidal anti-inflammatory drugs (NSAIDs). But the obvious side effects limit their use, so it is urgent to find new therapeutic targets. Postsynaptic density (PSD)-95 protein plays an important role in the occurrence and development of chronic pain. The over-expression of the PSD-95 protein and its interaction with other proteins are closely related to the chronic pain. Besides, the PSD-95-related drugs that inhibit the expression of PSD-95 as well as the interaction with other protein have been proved to treat chronic pain significantly. In conclusion, although more deep studies are needed in the future, these studies indicate that PSD-95 and the related proteins, such as NMDA receptor (NMDAR) subunit 2B (GluN2B), AMPA receptor (AMPAR), calmodulin-dependent protein kinase II (CaMKII), 5-hydroxytryptamine 2A receptor (5-HT2AR), and neuronal nitric oxide synthase (nNOS), are the promising therapeutic targets for chronic pain.
Collapse
Affiliation(s)
- Lulin Ma
- Department of Pain Medicine, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, China
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Dongdong Sun
- Hengyang Medical School, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Song Wen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Jie Yuan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Jing Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Xinran Tan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Song Cao
- Department of Pain Medicine, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, China.
| |
Collapse
|
2
|
do Nascimento AM, Marques RB, Roldão AP, Rodrigues AM, Eslava RM, Dale CS, Reis EM, Schechtman D. Exploring protein-protein interactions for the development of new analgesics. Sci Signal 2024; 17:eadn4694. [PMID: 39378285 DOI: 10.1126/scisignal.adn4694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024]
Abstract
The development of new analgesics has been challenging. Candidate drugs often have limited clinical utility due to side effects that arise because many drug targets are involved in signaling pathways other than pain transduction. Here, we explored the potential of targeting protein-protein interactions (PPIs) that mediate pain signaling as an approach to developing drugs to treat chronic pain. We reviewed the approaches used to identify small molecules and peptide modulators of PPIs and their ability to decrease pain-like behaviors in rodent animal models. We analyzed data from rodent and human sensory nerve tissues to build associated signaling networks and assessed both validated and potential interactions and the structures of the interacting domains that could inform the design of synthetic peptides and small molecules. This resource identifies PPIs that could be explored for the development of new analgesics, particularly between scaffolding proteins and receptors for various growth factors and neurotransmitters, as well as ion channels and other enzymes. Targeting the adaptor function of CBL by blocking interactions between its proline-rich carboxyl-terminal domain and its SH3-domain-containing protein partners, such as GRB2, could disrupt endosomal signaling induced by pain-associated growth factors. This approach would leave intact its E3-ligase functions, which are mediated by other domains and are critical for other cellular functions. This potential of PPI modulators to be more selective may mitigate side effects and improve the clinical management of pain.
Collapse
Affiliation(s)
- Alexandre Martins do Nascimento
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
- Laboratory of Neuromodulation of Experimental Pain (LaNed), Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, SP 05508-000, Brazil
| | - Rauni Borges Marques
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
- Interunit Graduate Program in Bioinformatics, University of São Paulo, SP 05508-000, Brazil
| | - Allan Pradelli Roldão
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Ana Maria Rodrigues
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Rodrigo Mendes Eslava
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Camila Squarzoni Dale
- Laboratory of Neuromodulation of Experimental Pain (LaNed), Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, SP 05508-000, Brazil
| | - Eduardo Moraes Reis
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Deborah Schechtman
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| |
Collapse
|
3
|
Shi YQ, Sun ZH, Wang ZZ, Su CY, Zhang W, Yu LY, Xu Y, Gao YL, Wang HB, Tian JW, Li CM. A novel role for microtubule affinity-regulating kinases in neuropathic pain. Br J Pharmacol 2024; 181:2012-2032. [PMID: 38112022 DOI: 10.1111/bph.16303] [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: 08/03/2023] [Revised: 09/15/2023] [Accepted: 10/03/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND AND PURPOSE Neuropathic pain affects millions of patients, but there are currently few viable therapeutic options available. Microtubule affinity-regulating kinases (MARKs) regulate the dynamics of microtubules and participate in synaptic remodelling. It is unclear whether these changes are involved in the central sensitization of neuropathic pain. This study examined the role of MARK1 or MARK2 in regulating neurosynaptic plasticity induced by neuropathic pain. EXPERIMENTAL APPROACH A rat spinal nerve ligation (SNL) model was established to induce neuropathic pain. The role of MARKs in nociceptive regulation was assessed by genetically knocking down MARK1 or MARK2 in amygdala and systemic administration of PCC0105003, a novel small molecule MARK inhibitor. Cognitive function, anxiety-like behaviours and motor coordination capability were also examined in SNL rats. Synaptic remodelling-associated signalling changes were detected with electrophysiological recording, Golgi-Cox staining, western blotting and qRT-PCR. KEY RESULTS MARK1 and MARK2 expression levels in amygdala and spinal dorsal horn were elevated in SNL rats. MARK1 or MARK2 knockdown in amygdala and PCC0105003 treatment partially attenuated pain-like behaviours along with improving cognitive deficit, anxiogenic-like behaviours and motor coordination in SNL rats. Inhibition of MARKs signalling reversed synaptic plasticity at the functional and structural levels by suppressing NR2B/GluR1 and EB3/Drebrin signalling pathways both in amygdala and spinal dorsal horn. CONCLUSION AND IMPLICATIONS These results suggest that MARKs-mediated synaptic remodelling plays a key role in the pathogenesis of neuropathic pain and that pharmacological inhibitors of MARKs such as PCC0105003 could represent a novel therapeutic strategy for the management of neuropathic pain.
Collapse
Affiliation(s)
- Yao-Qin Shi
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| | - Zhi-Hong Sun
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| | - Zhe-Zhe Wang
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| | - Chun-Yu Su
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| | - Wei Zhang
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| | - Lin-Yao Yu
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| | - Yang Xu
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| | - Yong-Lin Gao
- College of Life Sciences, Yantai University, Yantai, China
| | - Hong-Bo Wang
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| | - Jing-Wei Tian
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| | - Chun-Mei Li
- From the school of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong Province, China
| |
Collapse
|
4
|
Wu J, Jin M, Tran Q, Kim M, Kim SI, Shin J, Park H, Shin N, Kang H, Shin HJ, Lee SY, Cui SB, Lee CJ, Lee WH, Kim DW. Employing the sustained-release properties of poly(lactic-co-glycolic acid) nanoparticles to reveal a novel mechanism of sodium-hydrogen exchanger 1 in neuropathic pain. Transl Res 2024; 263:53-72. [PMID: 37678757 DOI: 10.1016/j.trsl.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/16/2023] [Accepted: 09/01/2023] [Indexed: 09/09/2023]
Abstract
Neuropathic pain is caused by injury or disease of the somatosensory system, and its course is usually chronic. Several studies have been dedicated to investigating neuropathic pain-related targets; however, little attention has been paid to the persistent alterations that these targets, some of which may be crucial to the pathophysiology of neuropathic pain. The present study aimed to identify potential targets that may play a crucial role in neuropathic pain and validate their long-term impact. Through bioinformatics analysis of RNA sequencing results, we identified Slc9a1 and validated the reduced expression of sodium-hydrogen exchanger 1 (NHE1), the protein that Slc9a1 encodes, in the spinal nerve ligation (SNL) model. Colocalization analysis revealed that NHE1 is primarily co-localized with vesicular glutamate transporter 2-positive neurons. In vitro experiments confirmed that poly(lactic-co-glycolic acid) nanoparticles loaded with siRNA successfully inhibited NHE1 in SH-SY5Y cells, lowered intracellular pH, and increased intracellular calcium concentrations. In vivo experiments showed that sustained suppression of spinal NHE1 expression by siRNA-loaded nanoparticles resulted in delayed hyperalgesia in naïve and SNL model rats, whereas amiloride-induced transient suppression of NHE1 expression yielded no significant changes in pain sensitivity. We identified Slc9a1, which encodes NHE1, as a key gene in neuropathic pain. Utilizing the sustained release properties of nanoparticles enabled us to elucidate the chronic role of decreased NHE1 expression, establishing its significance in the mechanisms of neuropathic pain.
Collapse
Affiliation(s)
- Junhua Wu
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Neurology, Yanji Hospital, Yanji, China
| | - Meiling Jin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Quangdon Tran
- Molecular Biology Laboratory, Department of Medical Laboratories, Hai Phong International Hospital, Hai Phong City, Vietnam
| | - Minwoo Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Nara Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyunji Kang
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sun Yeul Lee
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Song-Biao Cui
- Department of Neurology, Affiliated Hospital of Yanbian University, Yanji, China
| | - C Justin Lee
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Won Hyung Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
5
|
Chen W, Jiang B, Zhao Y, Yu W, Zhang M, Liang Z, Liu X, Ye B, Chen D, Yang L, Li F. Discovery of benzyloxy benzamide derivatives as potent neuroprotective agents against ischemic stroke. Eur J Med Chem 2023; 261:115871. [PMID: 37852031 DOI: 10.1016/j.ejmech.2023.115871] [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: 07/13/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Aberrant activation of N-methyl-d-aspartate receptors (NMDAR) and the resulting neuronal nitric oxide synthase (nNOS) excessive activation play crucial pathogenic roles in neuronal damage caused by stroke. Disrupting postsynaptic density protein 95 (PSD95)-nNOS protein-protein interaction (PPI) has been proposed as a potential therapeutic strategy for ischemic stroke without incurring the unwanted side effects of direct NMDAR antagonism. Based on a specific PSD95-nNOS PPI inhibitor (SCR4026), we conducted a detailed study on structure-activity relationship (SAR) to discover a series of novel benzyloxy benzamide derivatives. Here, our efforts resulted in the best 29 (LY836) with improved neuroprotective activities in primary cortical neurons from glutamate-induced damage and drug-like properties. Whereafter, co-immunoprecipitation experiment demonstrated that 29 significantly blocked PSD95-nNOS association in cultured cortical neurons. Furthermore, 29 displayed good pharmacokinetic properties (T1/2 = 4.26 and 4.08 h after oral and intravenous administration, respectively) and exhibited powerful therapeutic effects in rats subjected to middle cerebral artery occlusion (MCAO) by reducing infarct size and neurological deficit score. These findings suggested that compound 29 may be a promising neuroprotection agent for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Weilin Chen
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Bo Jiang
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yifan Zhao
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Wei Yu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Minyue Zhang
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Zhenchu Liang
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xing Liu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Binglin Ye
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Dongyin Chen
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Lei Yang
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Fei Li
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
6
|
Liu YJ, Li YL, Fang ZH, Liao HL, Zhang YY, Lin J, Liu F, Shen JF. NMDARs mediate peripheral and central sensitization contributing to chronic orofacial pain. Front Cell Neurosci 2022; 16:999509. [PMID: 36238833 PMCID: PMC9553029 DOI: 10.3389/fncel.2022.999509] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Peripheral and central sensitizations of the trigeminal nervous system are the main mechanisms to promote the development and maintenance of chronic orofacial pain characterized by allodynia, hyperalgesia, and ectopic pain after trigeminal nerve injury or inflammation. Although the pathomechanisms of chronic orofacial pain are complex and not well known, sufficient clinical and preclinical evidence supports the contribution of the N-methyl-D-aspartate receptors (NMDARs, a subclass of ionotropic glutamate receptors) to the trigeminal nociceptive signal processing pathway under various pathological conditions. NMDARs not only have been implicated as a potential mediator of pain-related neuroplasticity in the peripheral nervous system (PNS) but also mediate excitatory synaptic transmission and synaptic plasticity in the central nervous system (CNS). In this review, we focus on the pivotal roles and mechanisms of NMDARs in the trigeminal nervous system under orofacial neuropathic and inflammatory pain. In particular, we summarize the types, components, and distribution of NMDARs in the trigeminal nervous system. Besides, we discuss the regulatory roles of neuron-nonneuronal cell/neuron-neuron communication mediated by NMDARs in the peripheral mechanisms of chronic orofacial pain following neuropathic injury and inflammation. Furthermore, we review the functional roles and mechanisms of NMDARs in the ascending and descending circuits under orofacial neuropathic and inflammatory pain conditions, which contribute to the central sensitization. These findings are not only relevant to understanding the underlying mechanisms, but also shed new light on the targeted therapy of chronic orofacial pain.
Collapse
Affiliation(s)
- Ya-Jing Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue-Ling Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhong-Han Fang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hong-Lin Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiu Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie-Fei Shen Fei Liu
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie-Fei Shen Fei Liu
| |
Collapse
|
7
|
Postsynaptic signaling at glutamatergic synapses as therapeutic targets. Curr Opin Neurobiol 2022; 75:102585. [PMID: 35738196 DOI: 10.1016/j.conb.2022.102585] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/29/2022] [Accepted: 05/17/2022] [Indexed: 11/21/2022]
Abstract
Dysregulation of glutamatergic synapses plays an important role in the pathogenesis of neurological diseases. In addition to mediating excitatory synaptic transmission, postsynaptic glutamate receptors interact with various membrane and intracellular proteins. They form structural and/or signaling synaptic protein complexes and thereby play diverse postsynaptic functions. Recently, several postsynaptic protein complexes have been associated with various neurological diseases and hence, have been characterized as important therapeutic targets. Moreover, novel small molecules and therapeutic peptides targeting and modulating the activities of these protein complexes have been discovered, some of which have advanced through preclinical translational research and/or clinical studies. This article describes the recent investigation of eight key protein complexes associated with the postsynaptic ionotropic and metabotropic glutamate receptors as therapeutic targets for central nervous system diseases.
Collapse
|