1
|
Li P, Wang J, Wang M, Chen X, Zhu H, Dong M. Development of GluN2A NMDA receptor positive allosteric modulators: Recent advances and perspectives. Bioorg Med Chem 2025; 124:118194. [PMID: 40239379 DOI: 10.1016/j.bmc.2025.118194] [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: 01/09/2025] [Revised: 03/17/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
N-methyl-d-aspartate (NMDA) receptors, functioning as glutamate-gated ion channels, mediate the permeation of Ca2+ and are essential for excitatory synaptic transmission and synaptic plasticity within the central nervous system (CNS). During brain development, there is a switch from an early dominance of GluN2B subunit expression to the incorporation of GluN2A subunits at mature synapses. NMDARs hypofunction is implicated in various psychiatric disorders, and activation of NMDARs containing GluN2A has recently attracted attention as a promising therapeutic approach for treating these diseases. This review focuses on the selective positive allosteric modulators (PAMs) that specifically target the ligand-binding domain (LBD) and N-terminal domain (NTD) regions of GluN2A subtype, as well as non-subunit selective PAMs, and discusses their implications in neuropsychiatric diseases such as stroke, depression, Alzheimer's disease, and Huntington's disease.
Collapse
Affiliation(s)
- Ping Li
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Jiacheng Wang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Mengjiao Wang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Xin Chen
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao 266021, China; National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hongyu Zhu
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Mingxin Dong
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
2
|
Gong H, Xu X, Talifu Z, Zhang CJ, Sun YZ, Yue ZM, Rao JS, Du LJ, Du XX. Prospects and challenges in NMDAR signaling in spinal cord injury recovery and neural circuit remodeling. Regen Ther 2025; 29:381-389. [PMID: 40265135 PMCID: PMC12013404 DOI: 10.1016/j.reth.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
N-methyl-d-aspartate receptors (NMDARs) are essential for excitatory synaptic transmission in the central nervous system, contributing to various physiological and pathological functions including learning, memory, neural development, synaptic transmission, and plasticity. NMDAR signaling plays a role in spinal cord injury outcomes, including restoring spinal circuits, modulating synaptic plasticity, reinstating synchronized functions, enhancing motor capabilities, and reducing neuropathic pain. Consequently, targeting NMDARs may serve as a promising approach to enhance axonal regeneration and reorganization of neural circuits following spinal injury.
Collapse
Affiliation(s)
- Han Gong
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Beijing International Cooperation Bases for Science and Technology on Biomaterials and Neural Regeneration, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
- University of Health and Rehabilitation Sciences, Qingdao City, 266113, China
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| | - Xin Xu
- University of Health and Rehabilitation Sciences, Qingdao City, 266113, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250100, China
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, 250100, China
| | - Zuliyaer Talifu
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Chun-Jia Zhang
- Department of Rehabilitation Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yu-Zhe Sun
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| | - Zhao-Ming Yue
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| | - Jia-Sheng Rao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Beijing International Cooperation Bases for Science and Technology on Biomaterials and Neural Regeneration, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | - Liang-Jie Du
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| | - Xiao-Xia Du
- University of Health and Rehabilitation Sciences, Qingdao City, 266113, China
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| |
Collapse
|
3
|
Rodrigues LC, Godoi ABD, Fais VC, Peterson RT, Maurer-Morelli CV, Costa JL. Zebrafish embryo-larval testing reveals differential toxicity of new psychoactive substances. Toxicol Rep 2025; 14:102018. [PMID: 40235715 PMCID: PMC11999215 DOI: 10.1016/j.toxrep.2025.102018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/03/2025] [Accepted: 03/29/2025] [Indexed: 04/17/2025] Open
Abstract
New psychoactive substances (NPS) have emerged as a significant public health concern, with synthetic cannabinoid receptor agonists (SCRAs) and ketamine derivatives being among the most frequently detected compounds in the forensic context worldwide. The Fish Embryo Acute Toxicity (FET) and Maximum Tolerated Concentration (MTC) tests are used to evaluate the acute toxicity of chemicals. In this study, we used these assays to evaluate the acute toxicity of three NPS in zebrafish embryos and larvae: the SCRA MDMB-4en-PINACA and the ketamine derivatives deschloroketamine (DCK) and 2-fluorodeschloroketamine (2F-DCK). Our findings demonstrated that MDMB-4en-PINACA induced severe developmental abnormalities, including pericardial edema and yolk edema, along with high embryo mortality (10 µM), characterized by endpoints such as coagulation, lack of heartbeat, and lack of somite formation. In contrast, DCK and 2F-DCK exhibited low embryo mortality even at higher concentrations. In larval stages, MDMB-4en-PINACA presented 8 % larvae mortality (10 µM) at eight days post-fertilization (dpf), whereas ketamine derivatives led to 100 % mortality at 2000 µM in the MTC test at eight dpf. The LC50 was calculated for the FET test with MDMB-4en-PINACA, and MTC test for both DCK and 2F-DCK. Additionally, our results support the absence of N-methyl-D-aspartate (NMDA) receptors in the early life stages of zebrafish described in previous studies and highlight the significance of ketamine derivatives intoxications when the NMDA receptor is expressed. Notably, MDMB-4en-PINACA exhibited significantly higher toxicity, with an LC50 of approximately 26 times lower than that of the ketamine derivatives. These results are particularly relevant given the increasing global prevalence of NPS-related intoxications and fatalities. Using zebrafish as an in vivo model for toxicological research provides an efficient approach for screening the acute effects of emerging compounds such as NPS.
Collapse
Affiliation(s)
- Leonardo Costalonga Rodrigues
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-887, Brazil
- Campinas Poison Control Center, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-881, Brazil
- Laboratory of Zebrafish, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-887, Brazil
| | - Alexandre Barcia de Godoi
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-887, Brazil
- Campinas Poison Control Center, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-881, Brazil
| | - Viviane Cristina Fais
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-887, Brazil
- Laboratory of Zebrafish, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-887, Brazil
| | | | - Claudia Vianna Maurer-Morelli
- School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-887, Brazil
- Laboratory of Zebrafish, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-887, Brazil
| | - Jose Luiz Costa
- Campinas Poison Control Center, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-881, Brazil
- Faculty of Pharmaceutical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, Sao Paulo 13083-871, Brazil
| |
Collapse
|
4
|
Forouzanfar F, Ahmadzadeh AM, Pourbagher-Shahri AM, Gorji A. Significance of NMDA receptor-targeting compounds in neuropsychological disorders: An in-depth review. Eur J Pharmacol 2025; 999:177690. [PMID: 40315950 DOI: 10.1016/j.ejphar.2025.177690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/16/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
N-methyl-D-aspartate receptors (NMDARs), a subclass of glutamate-gated ion channels, play an integral role in the maintenance of synaptic plasticity and excitation-inhibition balance within the central nervous system (CNS). Any irregularities in NMDAR functions, whether hypo-activation or over-activation, can destabilize neural networks and impair CNS function. Several decades of experimental and clinical investigations have demonstrated that NMDAR dysfunction is implicated in the pathophysiology of various neurological disorders. Despite designing a long list of compounds that differentially modulate NMDARs, success in developing drugs that can selectively and effectively regulate various NMDAR subtypes while showing encouraging efficacy in clinical settings remains limited. A better understanding of the basic mechanism of NMDAR function, particularly its selective regulation in pathological conditions, could aid in designing effective drugs for the treatment of neurological conditions. Here, we reviewed the experimental and clinical investigations that studied the effects of available NMDAR modulators in various neurological disorders and weighed up the pros and cons of the use of these substances on the improvement of functional outcomes of these disorders. Despite numerous efforts to develop NMDAR modulatory drugs that did not produce the desired outcomes, NMDARs remain a significant target for advancing novel drugs to treat neurological disorders. This article reviews the complexity of NMDAR signaling dysfunction in different neurological diseases, the efforts taken to examine designed compounds targeting specific subtypes of NMDARs, including challenges associated with using these substances, and the potential enhancements in drug discovery for NMDAR modulatory compounds by innovative technologies.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Mahmoud Ahmadzadeh
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Radiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Mohammad Pourbagher-Shahri
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran; Department of Neurosurgery, Münster University, Münster, Germany; Epilepsy Research Center, Münster University, Münster, Germany.
| |
Collapse
|
5
|
Youn T, Kim G, Hariharan P, Li X, Ahmed W, Byrne B, Liu X, Guan L, Chae PS. Improved Pendant-Bearing Glucose-Neopentyl Glycols for Membrane Protein Stability. Bioconjug Chem 2025; 36:707-717. [PMID: 40105011 DOI: 10.1021/acs.bioconjchem.4c00556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Membrane proteins are biologically and pharmaceutically significant, and determining their 3D structures requires a membrane-mimetic system to maintain protein stability. Detergent micelles are widely used as membrane mimetics; however, their dynamic structures often lead to the denaturation and aggregation of encapsulated membrane proteins. To address the limitations of classical detergents in stabilizing membrane proteins, we previously reported a class of glucose-neopentyl glycols (GNGs) and their pendant-bearing versions (P-GNGs), several of which proved more effective than DDM in stabilizing membrane proteins. In this study, we synthesized additional GNG derivatives by varying the lengths of the pendant (P-GNGs), and by introducing hemifluorinated pendants to the GNG scaffold (fluorinated pendant-bearing GNGs or FP-GNGs). The synthetic flexibility of the GNG chemical architecture allowed us to create a diverse range of derivatives, essential for the effective optimization of detergent properties. When tested with two model membrane proteins (a transporter and a G-protein coupled receptor (GPCR)), most of the new (F)P-GNGs demonstrated superior stabilization of these membrane proteins compared to DDM, the original GNG (OGNG)), and a previously developed P-GNG (i.e., GNG-3,14). Notably, several P-GNGs synthesized in this study were as effective as or even better than lauryl maltose neopentyl glycol (LMNG) in stabilizing a human GPCR, beta2 adrenergic receptor (β2AR). Enhanced protein stability was particularly observed for the P-GNGs with a butyl (C4) or pentyl (C5) pendant, indicating that these pendant sizes are optimal for membrane protein stability. The volumes of these pendants appear to minimize the empty spaces in the micelle interiors, thereby enhancing detergent-detergent interactions in micelles complexed with the membrane proteins. Additionally, we identified one FP-GNG that was more efficient at extracting the transporter and more effective at stabilizing the GPCR than DDM. Thus, the current study demonstrates that both chain length and number of fluorine atoms in the pendants of the P-GNGs were crucial determinants for membrane protein stability. We not only developed a few (F)P-GNGs that are significantly more effective than maltoside detergents (LMNG/DDM) for protein extraction and stability but we also provided an effective strategy for detergent design through the utilization of partially fluorinated pendants of varying length.
Collapse
Affiliation(s)
- Taeyeol Youn
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, South Korea
| | - Ganghee Kim
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, South Korea
| | - Parameswaran Hariharan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Xianglan Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Waqar Ahmed
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, South Korea
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
| | - Xiangyu Liu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Lan Guan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Pil Seok Chae
- Department of Bionano Engineering, Hanyang University ERICA, Ansan 155-88, South Korea
| |
Collapse
|
6
|
Huang X, Sun X, Wang Q, Zhang J, Wen H, Chen WJ, Zhu S. Structural insights into the diverse actions of magnesium on NMDA receptors. Neuron 2025; 113:1006-1018.e4. [PMID: 40010346 DOI: 10.1016/j.neuron.2025.01.021] [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: 08/23/2024] [Revised: 12/09/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025]
Abstract
Magnesium (Mg2+) is a key regulatory ion of N-methyl-ᴅ-aspartate (NMDA) receptors, including conferring them to function as coincidence detectors for excitatory synaptic transmission. However, the structural basis underlying the Mg2+ action on NMDA receptors remains unclear. Here, we report the cryo-EM structures of GluN1-N2B receptors and identify three distinct Mg2+-binding pockets. Specifically, site Ⅰ is located at the selectivity filter where an asparagine ring forms coordination bonds with Mg2+ and is responsible for the voltage-dependent block. Sites Ⅱ and Ⅲ are located at the N-terminal domain (NTD) of the GluN2B subunit and involved in the allosteric potentiation and inhibition, respectively. Site Ⅱ consists of three acidic residues, and the combination of three mutations abolishes the GluN2B-specific Mg2+ potentiation, while site Ⅲ overlaps with the Zn2+ pocket, and mutations here significantly reduce the inhibition. Our study enhances the understanding of multifaceted roles of Mg2+ in NMDA receptors and synaptic plasticity.
Collapse
Affiliation(s)
- Xuejing Huang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Xiaole Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Jilin Zhang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Han Wen
- DP Technology, Beijing 100089, China; AI for Science Institute, Beijing 100085, China; State Key Laboratory of Medical Proteomics, Beijing 102206, China
| | - Wan-Jin Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian 350005, China.
| | - Shujia Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
7
|
Kang H, Epstein M, Banke TG, Perszyk R, Simorowski N, Paladugu S, Liotta DC, Traynelis SF, Furukawa H. Structural basis for channel gating and blockade in tri-heteromeric GluN1-2B-2D NMDA receptor. Neuron 2025; 113:991-1005.e5. [PMID: 39954679 PMCID: PMC11968220 DOI: 10.1016/j.neuron.2025.01.013] [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: 10/01/2024] [Revised: 12/09/2024] [Accepted: 01/21/2025] [Indexed: 02/17/2025]
Abstract
Discrete activation of N-methyl-D-aspartate receptor (NMDAR) subtypes by glutamate and the co-agonist glycine is fundamental to neuroplasticity. A distinct variant, the tri-heteromeric receptor, comprising glycine-binding GluN1 and two types of glutamate-binding GluN2 subunits, exhibits unique pharmacological characteristics, notably enhanced sensitivity to the anti-depressant channel blocker S-(+)-ketamine. Despite its significance, the structural mechanisms underlying ligand gating and channel blockade of tri-heteromeric NMDARs remain poorly understood. Here, we identify and characterize tri-heteromeric GluN1-2B-2D NMDAR in the adult brain, resolving its structures in the activated, inhibited, and S-(+)-ketamine-blocked states. These structures reveal the ligand-dependent conformational dynamics that modulate the tension between the extracellular domain and transmembrane channels, governing channel gating and blockade. Additionally, we demonstrate that the inhibitor (S)-DQP-997-74 selectively decouples linker tension in GluN2D, offering insights into subtype-selective targeting for cognitive modulation.
Collapse
Affiliation(s)
- Hyunook Kang
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Max Epstein
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Tue G Banke
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Riley Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Noriko Simorowski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Srinu Paladugu
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | | | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
8
|
Bao N, Liu J, Wang H, Xing L, Xie Z, Liu C, Jin S, Jia J, Zhang M, Fan J. Drug Repurposing and Screening for Multiple Sclerosis Targeting Microglia and Macrophages. Mol Neurobiol 2025; 62:4724-4742. [PMID: 39485630 DOI: 10.1007/s12035-024-04602-w] [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: 04/11/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Microglia/macrophages (MG/Mφ) play a central role in the pathogenesis of multiple sclerosis (MS). However, the intricacies of the immunomodulatory microenvironment in MS, particularly the heterogeneity and regulatory mechanisms of MG/Mφ subpopulations, remain elusive. The commonly used treatment options for MS have several drawbacks, such as significant side effects and uncertain efficacy. The exploration of developing new drugs targeting MG/Mφ for the treatment of MS remains to be investigated. We identified three distinct subpopulations of MG/Mφ, among which MG/Mφ_3 significantly increased as the experimental autoimmune encephalomyelitis (EAE) progressed. Ifenprodil and RO-25-6981 demonstrated notable inhibition of inflammatory factor expression, accompanied by reduced cytotoxicity. The interaction modes of these compounds with the common binding pocket in the GluN1b-GluN2B amino terminal domain heterodimer were elucidated. Virtual docking, based on the N-methyl-D-aspartate (NMDA) receptor, showed that homo-skeleton compounds of ifenprodil potentially exhibit low binding free energy with the receptor, including eliprodil and volinanserin. In vitro cell models corroborated the effective inhibition of inflammatory factor expression and minimal cytotoxicity of eliprodil and volinanserin. CoMFA (standard error of estimate = 0.378, R2 = 0.928, F values = 241.255, Prob. of R2 = 0) and topomer CoMFA (q2 = 0.553, q2 stderr = 0.77, intercept = - 1.48, r2 = 0.908, r2 stderr = 0.35) were established based on the inhibitors of NMDA receptor. The contour maps of CoMFA and topomer CoMFA models give structural information to improve the inhibitory function. This study underscores the involvement of MG/Mφ in inflammatory pathways during MS progression and offers promising compound candidates for MS therapy targeting MG/Mφ.
Collapse
Affiliation(s)
- Nandi Bao
- Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Jing Liu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Heran Wang
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Lei Xing
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Zhonghui Xie
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chuanbin Liu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Shaowei Jin
- National Supercomputing Shenzhen Center, Shenzhen, 518052, China
| | - Jianjun Jia
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Minghua Zhang
- Medical Supplies Center of PLA General Hospital, Beijing, 100853, China.
| | - Jiao Fan
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
9
|
Wang ZC, Zeng Y, Sun JY, Chen XQ, Wu HC, Li YY, Mu YG, Zheng LZ, Gao ZB, Li WF. An efficient deep learning-based strategy to screen inhibitors for GluN1/GluN3A receptor. Acta Pharmacol Sin 2025:10.1038/s41401-025-01513-x. [PMID: 40069493 DOI: 10.1038/s41401-025-01513-x] [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: 12/02/2024] [Accepted: 02/12/2025] [Indexed: 03/15/2025]
Abstract
The GluN1/GluN3A receptor, a unique excitatory glycine receptor recently identified in the central nervous system, challenges traditional perspectives of N-methyl-D-aspartate (NMDA) receptor diversity and glycinergic signaling. Its role in emotional regulation positions it as a potential therapeutic target for neuropsychiatric disorders. However, pharmacological research on GluN1/GluN3A receptors remains at an early stage. Traditional high-throughput screening methods for ion channel drug discovery often lack efficiency, particularly when applied to large compound libraries. To address this concern, we designed a deep learning-based strategy that balances efficiency and accuracy for identifying GluN1/GluN3A inhibitors. First, a sequence-based scoring function was developed to rapidly screen a library containing 18 million compounds, reducing the pool to approximately 105 candidates. Next, two complex-based scoring functions, IGModel and RTMScore, were employed to precisely score and rank the remaining candidates. Finally, an active molecule with an IC50 of 2.87 ± 0.80 μM for the GluN1/GluN3A receptor was confirmed through whole-cell voltage-clamp electrophysiology. This study also presents a paradigm for integrating deep learning into rapid and precise high-throughput screening.
Collapse
Affiliation(s)
- Ze-Chen Wang
- School of Physics, Shandong University, Jinan, 250100, China
| | - Yue Zeng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 200032, China
| | - Jin-Yuan Sun
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue-Qin Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hao-Chen Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yang-Yang Li
- School of Physics, Shandong University, Jinan, 250100, China
| | - Yu-Guang Mu
- School of Biological Science, Nanyang Technological University, Singapore, 637551, Singapore
| | - Liang-Zhen Zheng
- Shenzhen Zelixir Biotech Co. Ltd, Hengtaiyu Park, Shenzhen, 518107, China
| | - Zhao-Bing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Wei-Feng Li
- School of Physics, Shandong University, Jinan, 250100, China.
| |
Collapse
|
10
|
Chen Y, Yue X, Tang Y, Zhu Q, Yu W, Luo M, Huang Y, Wen L, Li F. Thrombin Nanochannel Logic Gate Inspired by BioMemory. Anal Chem 2025; 97:3220-3226. [PMID: 39620937 DOI: 10.1021/acs.analchem.4c02983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
The process of "reading" and "writing" in biomemory involves the transmission of electrical signals between neurons, with ligand-gated ion channels assuming a key role. The solid-state nanochannels exhibit certain similarities with neurons. Information transmission can be achieved by controlling the flow of ions within nanochannels, rendering them potentially suitable for simulating neuron behavior. Herein, thrombin (Thr) was chosen as the target protein, and a functionalized nanochannel sensing system was successfully constructed using DNA aptamers, enabling a highly sensitive Thr response with a detection limit of 0.221 fM. Simultaneously, based on Watson-Crick base pairing and programmable chain displacement reactions, controlled release and cyclic response of the target molecule were further achieved. This mechanism elucidates the rules governing specific input-output relationships, innovatively linking them with memory storage and recognition through the Thr-nanochannel logic gate, thereby realizing the reading of biomemory at the hardware level. In summary, the biological hybrid nanofluidic control device of this invention converts molecular events into electrical signals, providing potential avenues for establishing connections between the mechanisms of biomemory and solid-state nanochannel biosensing and recognition in the future.
Collapse
Affiliation(s)
- Yonghuan Chen
- College of Chemistry and Materials Science, Guangdong Provincial Key Laboratory of Speed Capability Research, Su Bingtian Center for Speed Research and Training, Jinan University, Guangzhou 510632, China
| | - Xinru Yue
- College of Chemistry and Materials Science, Guangdong Provincial Key Laboratory of Speed Capability Research, Su Bingtian Center for Speed Research and Training, Jinan University, Guangzhou 510632, China
| | - Yongtao Tang
- College of Chemistry and Materials Science, Guangdong Provincial Key Laboratory of Speed Capability Research, Su Bingtian Center for Speed Research and Training, Jinan University, Guangzhou 510632, China
| | - Qi Zhu
- College of Chemistry and Materials Science, Guangdong Provincial Key Laboratory of Speed Capability Research, Su Bingtian Center for Speed Research and Training, Jinan University, Guangzhou 510632, China
| | - Weihua Yu
- College of Chemistry and Materials Science, Guangdong Provincial Key Laboratory of Speed Capability Research, Su Bingtian Center for Speed Research and Training, Jinan University, Guangzhou 510632, China
| | - Mengfan Luo
- College of Chemistry and Materials Science, Guangdong Provincial Key Laboratory of Speed Capability Research, Su Bingtian Center for Speed Research and Training, Jinan University, Guangzhou 510632, China
| | - Yu Huang
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Material Science and Chemistry, China University of Geosciences, Wuhan 430074, P. R. China
| | - Liping Wen
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Fengyu Li
- College of Chemistry and Materials Science, Guangdong Provincial Key Laboratory of Speed Capability Research, Su Bingtian Center for Speed Research and Training, Jinan University, Guangzhou 510632, China
- College of Chemistry, Zhengzhou University, 450001 Zhengzhou, China
| |
Collapse
|
11
|
Capó T, Rebassa JB, Raïch I, Lillo J, Badia P, Navarro G, Reyes-Resina I. Future Perspectives of NMDAR in CNS Disorders. Molecules 2025; 30:877. [PMID: 40005187 PMCID: PMC11857888 DOI: 10.3390/molecules30040877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Neurodegenerative diseases such as Alzheimer's and Parkinson's diseases are among the leading causes of physical and cognitive disability across the globe. Fifty million people worldwide suffer these diseases, and that number is expected to rise as the population ages. Ictus is another pathology that also courses with neurodegeneration and is a leading cause of mortality and long-term disability in developed countries. Schizophrenia is not as common as other mental disorders, affecting approximately 24 million people worldwide. All these disorders have in common that still there is not an effective pharmacological treatment to cure them. The N-methyl-D-aspartate (NMDA) receptor (NMDAR) has attracted attention as a potential therapeutic target due to its important role in learning and memory and also due to its implication in excitotoxicity processes. Some drugs targeting NMDARs are already being used to treat symptoms of disorders affecting the central nervous system (CNS). Here, we aim to review the implications of NMDAR in these CNS pathologies, its role as a potential therapeutic target, and the future perspectives for developing new treatments focused on these receptors.
Collapse
Affiliation(s)
- Toni Capó
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| | - Joan Biel Rebassa
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| | - Iu Raïch
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| | - Jaume Lillo
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - Pau Badia
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
| | - Gemma Navarro
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| | - Irene Reyes-Resina
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (T.C.); (J.B.R.); (I.R.); (P.B.)
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Institute of Neuroscience, University of Barcelona (NeuroUB), Campus Mundet, Passeig de la Vall d’Hebron171, 08035 Barcelona, Spain
| |
Collapse
|
12
|
Zhao F, Atxabal U, Mariottini S, Yi F, Lotti JS, Layeux MS, Currier C, Maderia MP, Cornelison LE, Anderson CM, Schultz EP, Zhang Z, Jiang L, Gao Z, Liu N, Woodahl EL, Bunch L, Hansen KB, Clausen RP. Design of ( R)-3-(5-Thienyl)carboxamido-2-aminopropanoic Acid Derivatives as Novel NMDA Receptor Glycine Site Agonists: Variation in Molecular Geometry to Improve Potency and Augment GluN2 Subunit-Specific Activity. J Med Chem 2025; 68:3572-3590. [PMID: 39847708 PMCID: PMC11832032 DOI: 10.1021/acs.jmedchem.4c02715] [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: 11/06/2024] [Revised: 12/16/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025]
Abstract
NMDA receptor ligands have therapeutic potential in neurological and psychiatric disorders. We designed (R)-3-(5-thienyl)carboxamido-2-aminopropanoic acid derivatives with nanomolar agonist potencies at NMDA receptor subtypes (GluN12/A-D). These compounds are superagonists at GluN1/2C compared to glycine and partial to full agonists at GluN1/2A and GluN1/2D but display functional antagonism at GluN1/2B due to low agonist efficacy. Notably, 8d display 864% agonist efficacy at GluN1/2C relative to glycine, and 8j has high potency at GluN1/2A (0.018 μM), GluN1/2C (0.0029 μM), and GluN1/2D (0.016 μM). We evaluated the binding mode in the glycine site using molecular modeling and mutagenesis. In vitro absorption, distribution, metabolism, and excretion (ADME) assays predict high metabolic stability but poor blood-brain barrier permeability. However, an ester prodrug for the carboxylate group of 7j display moderately high blood-brain barrier permeability. The thiophenecarboxamide agonists expand the synthetic pharmacology of NMDA receptors and provide structural insights that facilitate the design of GluN1 agonists with GluN2 subunit-specific activity.
Collapse
Affiliation(s)
- Fabao Zhao
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry
of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong P. R. China
| | - Unai Atxabal
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Sofia Mariottini
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Feng Yi
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - James S. Lotti
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Michael S. Layeux
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Chandler Currier
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Matthew P. Maderia
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Lauren E. Cornelison
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Carly M. Anderson
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Eric P. Schultz
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Zhucheng Zhang
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Liyang Jiang
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry
of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong P. R. China
| | - Zhen Gao
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry
of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong P. R. China
| | - Na Liu
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry
of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong P. R. China
| | - Erica L. Woodahl
- L.S.
Skaggs Institute for Health Innovation, Department of Biomedical and
Pharmaceutical Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Lennart Bunch
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Kasper B. Hansen
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Rasmus P. Clausen
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| |
Collapse
|
13
|
Norris C, Murphy SF, VandeVord PJ. Acute astrocytic and neuronal regulation of glutamatergic protein expression following blast. Neurosci Lett 2025; 848:138108. [PMID: 39734031 DOI: 10.1016/j.neulet.2024.138108] [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: 09/07/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
Regulation of glutamate through glutamate-glutamine cycling is critical for mediating nervous system plasticity. Blast-induced traumatic brain injury (bTBI) has been linked to glutamate-dependent excitotoxicity, which may be potentiating chronic disorders such as post-traumatic epilepsy. The purpose of this study was to measure changes in the expression of astrocytic and neuronal proteins responsible for glutamatergic regulation at 4-, 12-, and 24 h in the cortex and hippocampus following single blast exposure in a rat model for bTBI. Animals were exposed to a blast with magnitudes ranging from 16 to 20 psi using an Advanced Blast Simulator, and western blotting was performed to compare changes in protein expression between blast and sham groups. Glial fibrillary acidic protein (GFAP) was increased at 24 h, consistent with astrocyte reactivity, yet no other proteins showed significant changes in expression at acute time points following blast (GS, GLT-1, GluN1, GluN2A, GluN2B). Therefore, these glutamate regulators likely do not play a major role in contributing to acute excitotoxicity or glial reactivity when analyzed by whole brain region. Investigation of substructural and subregional effects in future studies, particularly within the hippocampus (e.g., dentate gyrus, CA1, CA2, CA3), may reveal localized changes in expression and/or NMDAR subunit composition capable of potentiating bTBI molecular cascades. Nevertheless, alternative regulators are likely to demonstrate greater sensitivity as acute therapeutic targets contributing to bTBI pathophysiology following single blast exposure.
Collapse
Affiliation(s)
- Carly Norris
- School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Susan F Murphy
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA; Veterans Affairs Medical Center, Salem, VA, USA
| | - Pamela J VandeVord
- School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA; Veterans Affairs Medical Center, Salem, VA, USA.
| |
Collapse
|
14
|
He M, Wollmuth LP. Regulation of NMDAR activation efficiency by environmental factors and subunit composition. J Gen Physiol 2025; 157:e202413637. [PMID: 39576244 PMCID: PMC11586625 DOI: 10.1085/jgp.202413637] [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: 07/09/2024] [Revised: 09/28/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
NMDA receptors (NMDAR) convert the major excitatory neurotransmitter glutamate into a synaptic signal. A key question is how efficiently the ion channel opens in response to the rapid exposure to presynaptic glutamate release. Here, we applied glutamate to single channel outside-out patches and measured the successes of channel openings and the latency to first opening to assay the activation efficiency of NMDARs under different physiological conditions and with different human subunit compositions. For GluN1/GluN2A receptors, we find that various factors, including intracellular ATP and GTP, can enhance the efficiency of activation presumably via the intracellular C-terminal domain. Notably, an energy-based internal solution or increasing the time between applications to increase recovery time improved efficiency. However, even under these optimized conditions and with a 1-s glutamate application, there remained around 10-15% inefficiency. Channel activation became more inefficient with brief synaptic-like pulses of glutamate at 2 ms. Of the different NMDAR subunit compositions, GluN2B-containing NMDARs showed the lowest success rate and longest latency to first openings, highlighting that they display the most distinct activation mechanism. In contrast, putative triheteromeric GluN1/GluN2A/GluN2B receptors showed high activation efficiency. Despite the low open probability, NMDARs containing either GluN2C or GluN2D subunits displayed high activation efficiency, nearly comparable with that for GluN2A-containing receptors. These results highlight that activation efficiency in NMDARs can be regulated by environmental surroundings and varies across different subunits.
Collapse
Affiliation(s)
- Miaomiao He
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| | - Lonnie P. Wollmuth
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
15
|
Sheng XM, Guan W. GRIN2A and Schizophrenia: Scientific Evidence and Biological Mechanisms. Curr Neuropharmacol 2025; 23:621-634. [PMID: 39501956 DOI: 10.2174/011570159x327712241023084944] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/19/2024] [Accepted: 09/10/2024] [Indexed: 05/07/2025] Open
Abstract
Schizophrenia is a severe psychiatric disorder and a complex polygenic inherited disease that affects nearly 1% of the global population. Although considerable progress has been made over the past 10 years in the treatment of schizophrenia, antipsychotics are not universally effective and may have serious side effects. The hypofunction of glutamate NMDA receptors (NMDARs) in GABAergic interneurons has long been postulated to be the principal pathophysiology of schizophrenia. A recent study has shown that GRIN2A pathogenic variants are closely related to the aetiology of the disorder. GRIN2A encodes the GluN2A protein, which is a subunit of NMDAR. Most GRIN2A variants have been predicted to cause protein truncation, which results in reduced gene expression. Preclinical studies have indicated that GRIN2A mutations lead to NMDAR loss of function and substantially increase the risk of schizophrenia; however, their role in schizophrenia is not well understood. We hypothesise that the heterozygous loss of GRIN2A induces NMDAR hypofunction sufficient to confer a substantial risk of schizophrenia. Therefore, this review focuses on GRIN2A as a target for novel antipsychotics and discusses the mechanisms by which GRIN2A modulates antischizophrenic activities. Moreover, our review contributes to the understanding of the pathophysiology of schizophrenia to facilitate finding treatments for the cognitive and negative symptoms of schizophrenia.
Collapse
Affiliation(s)
- Xiao-Ming Sheng
- Department of Trauma Center, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, Jiangsu, China
| |
Collapse
|
16
|
Lotti JS, Jones J, Farnsworth JC, Yi F, Zhao F, Menniti FS, Volkmann RA, Clausen RP, Hansen KB. Evaluation of allosteric N-methyl-d-aspartate receptor modulation by GluN2A-selective antagonists using pharmacological equilibrium modeling. Mol Pharmacol 2025; 107:100004. [PMID: 39919165 DOI: 10.1124/molpharm.124.000975] [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: 06/20/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 01/22/2025] Open
Abstract
N-methyl-d-aspartate (NMDA)-type ionotropic glutamate receptors are critically involved in excitatory neurotransmission and their dysfunction is implicated in many brain disorders. Allosteric modulators with selectivity for specific NMDA receptor subtypes are therefore attractive as therapeutic agents, and sustained drug discovery efforts have resulted in a wide range of new allosteric modulators. However, evaluation of allosteric NMDA receptor modulators is limited by the lack of operational ligand-receptor models to describe modulator binding dissociation constants (KB) and effects on agonist binding affinity (α) and efficacy (β). Here, we describe a pharmacological equilibrium model that encapsulates activation and modulation of NMDA receptors, and we apply this model to afford deeper understanding of GluN2A-selective negative allosteric modulators, TCN-201, MPX-004, and MPX-007. We exploit slow negative allosteric modulator unbinding to examine receptors at hemi-equilibrium when fully occupied by agonists and modulators to demonstrate that TCN-201 display weaker binding and negative modulation of glycine binding affinity (KB = 42 nM, α = 0.0032) compared with MPX-004 (KB = 9.3 nM, α = 0.0018) and MPX-007 (KB = 1.1 nM, α = 0.00053). MPX-004 increases agonist efficacy (β = 1.19), whereas TCN-201 (β = 0.76) and MPX-007 (β = 0.82) reduce agonist efficacy. These values describing allosteric modulation of diheteromeric GluN1/2A receptors with 2 modulator binding sites are unchanged in triheteromeric GluN1/2A/2B receptors with a single binding site. This evaluation of NMDA receptor modulation reveals differences between ligand analogs that shape their utility as pharmacological tool compounds and facilitates the design of new modulators with therapeutic potential. SIGNIFICANCE STATEMENT: Detailed understanding of allosteric N-methyl-d-aspartate (NMDA) receptor modulation requires pharmacological methods to quantify modulator binding affinity and the strengths of modulation of agonist binding and efficacy. We describe a generic ligand-receptor model for allosteric NMDA receptor modulation and use this model for the characterization of GluN2A-selective negative allosteric modulators. The model enables quantitative evaluation of a broad range of NMDA receptor modulators and provides opportunities to optimize these modulators by embellishing the interpretation of their structure-activity relationships.
Collapse
Affiliation(s)
- James S Lotti
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana
| | - Jaron Jones
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana
| | - Jill C Farnsworth
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana
| | - Feng Yi
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Southern Medical University, Guangzhou, China
| | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Frank S Menniti
- MindImmune Therapeutics, Inc, George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
| | | | - Rasmus P Clausen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper B Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana.
| |
Collapse
|
17
|
Yang H, Kong L, Chen Z, Wu J. Effect of functional groups of polystyrene nanoplastics on the neurodevelopmental toxicity of acrylamide in the early life stage of zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 278:107177. [PMID: 39612669 DOI: 10.1016/j.aquatox.2024.107177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/17/2024] [Accepted: 11/24/2024] [Indexed: 12/01/2024]
Abstract
Polystyrene nanoplastics (PS NPs) and acrylamide (ACR), both emerging contaminants, have been found to be related to neurotoxicity. However, the effects of PS NPs on ACR-induced neurodevelopmental toxicity remain unclear. In this study, anionic carboxyl polystyrene nanoplastics (PS NPs-COOH), cationic amino polystyrene nanoplastics (PS NPs-NH2) and unmodified PS NPs were selected to investigate their interaction with ACR. A serious of the neurotoxicity biomarkers from individual to molecular level were evaluated to explore the specific mechanisms. The results indicated that the unmodified PS NPs had the most significant impact on embryonic development at low concentrations in combination with ACR. The toxicity of the other two functionalized PS NPs increased with concentration, exhibiting a clear dose-response relationship. Meanwhile, all three kinds of PS NPs significantly enhanced the impacts of ACR on the locomotion behavior of zebrafish larvae. Analysis of zebrafish nervous system development showed that PS NPs-COOH exhibit greater toxicity to the central nervous system. In contrast, PS NPs-NH2 had a more significant impact on the motor nervous system. Gene expression analysis revealed that ACR and PS NPs significantly affected the expression levels of neurodevelopmental related genes, including Neurog1, Elavl3, Gfap, Gap43, Mbpa, Shha. PS NPs modified with functional groups could induce corresponding neurotoxicity by affecting genes expression related to neuronal differentiation, motor neuron, and axonal development. Based on the comprehensive biomarker response index, the order of the impacts of NPs on the neurotoxicity of ACR was PS NPs-COOH > PS NPs-NH2 > PS NPs. In conclusion, this study provides new insights into the interactive biological effects of NPs and ACR on zebrafish embryo, contributing to a better understanding of their environmental risk to aquatic ecosystem.
Collapse
Affiliation(s)
- Haohan Yang
- College of Environmental Science and Engineering, Yangzhou University, Yangzhou 225127, China.
| | - Linghui Kong
- College of Environmental Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Zhuoyu Chen
- College of Environmental Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Jun Wu
- College of Environmental Science and Engineering, Yangzhou University, Yangzhou 225127, China
| |
Collapse
|
18
|
Rickgauer JP, Choi H, Moore AS, Denk W, Lippincott-Schwartz J. Structural dynamics of human ribosomes in situ reconstructed by exhaustive high-resolution template matching. Mol Cell 2024; 84:4912-4928.e7. [PMID: 39626661 DOI: 10.1016/j.molcel.2024.11.003] [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] [Revised: 07/29/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024]
Abstract
Protein synthesis is central to life and requires the ribosome, which catalyzes the stepwise addition of amino acids to a polypeptide chain by undergoing a sequence of structural transformations. Here, we employed high-resolution template matching (HRTM) on cryoelectron microscopy (cryo-EM) images of directly cryofixed living cells to obtain a set of ribosomal configurations covering the entire elongation cycle, with each configuration occurring at its native abundance. HRTM's position and orientation precision and ability to detect small targets (∼300 kDa) made it possible to order these configurations along the reaction coordinate and to reconstruct molecular features of any configuration along the elongation cycle. Visualizing the cycle's structural dynamics by combining a sequence of >40 reconstructions into a 3D movie readily revealed component and ligand movements, some of them surprising, such as spring-like intramolecular motion, providing clues about the molecular mechanisms involved in some still mysterious steps during chain elongation.
Collapse
Affiliation(s)
- J Peter Rickgauer
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Heejun Choi
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Andrew S Moore
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Winfried Denk
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | | |
Collapse
|
19
|
Mirmotahari SA, Aliomrani M, Hassanzadeh F, Sirous H, Rostami M. Hybrid derivatives containing dimethyl fumarate and benzothiazole scaffolds for the potential treatment of multiple sclerosis; in silico & in vivo study. Daru 2024; 32:599-615. [PMID: 39106020 PMCID: PMC11554962 DOI: 10.1007/s40199-024-00529-8] [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: 11/12/2022] [Accepted: 06/30/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Multiple Sclerosis (MS) is a chronic autoimmune, inflammatory neurological disease of the CNS. Riluzole and dimethyl fumarate (DMF) are two FDA-approved drugs to treat amyotrophic lateral sclerosis (ALS) and MS. Riluzole (a benzothiazole derivative) inhibits glutamate release from nerve terminals by antagonizing the N-Methyl-D-Aspartate (NMDA) receptor, and DMF upregulates anti-oxidative pathways. OBJECTIVES Herein, using molecular hybridization strategy, we synthesized some new hybrid structures of Riluzole and DMF through some common successive synthetic pathways for evaluating their potential activity for remyelination in MS treatment. METHODS Molecular docking experiments assessed the binding affinity of proposed structures to the NMDA active site. The designed structures were synthesized and purified based on well-known chemical synthesis procedures. Afterward, in vivo evaluation for their activity was done in the C57Bl/6 Cuprizone-induced demyelination MS model. RESULTS AND CONCLUSION The proposed derivatives were recognized to be potent enough based on docking studies (ΔGbind of all derivatives were -7.2 to -7.52 compare to the Ifenprodil (-6.98) and Riluzole (-4.42)). The correct structures of desired derivatives were confirmed using spectroscopic methods. Based on in vivo studies, D4 and D6 derivatives exhibited the best pharmacological results, although only D6 showed a statistically significant difference compared to the control. Also, for D4 and D6 derivatives, myelin staining confirmed reduced degeneration in the corpus callosum. Consequently, D4 and D6 derivatives are promising candidates for developing new NMDA antagonists with therapeutic value against MS disorders.
Collapse
Affiliation(s)
- Seyedeh Azin Mirmotahari
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R., Iran
| | - Mehdi Aliomrani
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Farshid Hassanzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R., Iran
| | - Hajar Sirous
- Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R., Iran
| | - Mahboubeh Rostami
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R., Iran.
| |
Collapse
|
20
|
Liu R, Liu N, Ma L, Liu Y, Huang Z, Peng X, Zhuang C, Niu J, Yu J, Du J. Research Progress on NMDA Receptor Enhancement Drugs for the Treatment of Depressive Disorder. CNS Drugs 2024; 38:985-1002. [PMID: 39379772 DOI: 10.1007/s40263-024-01123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2024] [Indexed: 10/10/2024]
Abstract
Major depressive disorder (MDD) is a severe mental illness with a complex etiology. Currently, many medications employed in clinical treatment exhibit limitations such as delayed onset of action and a high incidence of adverse reactions. Therefore, there is a pressing need to develop antidepressants that exhibit enhanced efficacy and safety. The N-methyl-D-aspartate receptor (NMDAR), a distinctive glutamate-gated ion channel receptor, has been implicated in the onset and progression of depressive disorder, as evidenced by both preclinical and clinical research. The NMDAR antagonist, ketamine, exhibits rapid and sustained antidepressant effects, holding promise as a novel therapeutic approach for depressive disorder. However, its psychotomimetic impact and potential for addiction have restricted its widespread clinical application. Notably, over the past decade, studies have suggested that enhancing NMDAR functionality can produce antidepressant effects with improved safety, especially with the emergence of NMDAR-positive allosteric modulators (PAMs). We view this as a potential novel strategy for treating depression, forming the basis for the narrative review that follows.
Collapse
Affiliation(s)
- Ruyun Liu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Ning Liu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Lin Ma
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Yue Liu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Zhuo Huang
- Department of Molecular and Cellular Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, Peking University School of Pharmaceutical Sciences, Beijing, China
| | - Xiaodong Peng
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jianguo Niu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China.
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China.
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China.
| | - Juan Du
- School of Pharmacy, Ningxia Medical University, 1160 Shengli St, Xingqing District, Yinchuan, Ningxia, China.
| |
Collapse
|
21
|
Michalski K, Abdulla T, Kleeman S, Schmidl L, Gómez R, Simorowski N, Vallese F, Prüss H, Heckmann M, Geis C, Furukawa H. Structural and functional mechanisms of anti-NMDAR autoimmune encephalitis. Nat Struct Mol Biol 2024; 31:1975-1986. [PMID: 39227719 PMCID: PMC11921143 DOI: 10.1038/s41594-024-01386-4] [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: 10/24/2023] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Autoantibodies against neuronal membrane proteins can manifest in autoimmune encephalitis, inducing seizures, cognitive dysfunction and psychosis. Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis is the most dominant autoimmune encephalitis; however, insights into how autoantibodies recognize and alter receptor functions remain limited. Here we determined structures of human and rat NMDARs bound to three distinct patient-derived antibodies using single-particle electron cryo-microscopy. These antibodies bind different regions within the amino-terminal domain of the GluN1 subunit. Through electrophysiology, we show that all three autoantibodies acutely and directly reduced NMDAR channel functions in primary neurons. Antibodies show different stoichiometry of binding and antibody-receptor complex formation, which in one antibody, 003-102, also results in reduced synaptic localization of NMDARs. These studies demonstrate mechanisms of diverse epitope recognition and direct channel regulation of anti-NMDAR autoantibodies underlying autoimmune encephalitis.
Collapse
Affiliation(s)
- Kevin Michalski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Taha Abdulla
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Sam Kleeman
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Lars Schmidl
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Ricardo Gómez
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Noriko Simorowski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Francesca Vallese
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Manfred Heckmann
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Christian Geis
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
- School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
22
|
Bej A, Hell JW, Ames JB. Chemical shift assignments of the α-actinin C-terminal EF-hand domain bound to a cytosolic C0 domain of GluN1 (residues 841-865) from the NMDA receptor. BIOMOLECULAR NMR ASSIGNMENTS 2024; 18:239-244. [PMID: 39207574 PMCID: PMC11511685 DOI: 10.1007/s12104-024-10194-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
N-methyl-D-aspartate receptors (NMDARs) consist of glycine-binding GluN1 and glutamate-binding GluN2 subunits that form tetrameric ion channels. NMDARs in the brain are important for controlling neuronal excitability to promote synaptic plasticity. The cytoskeletal protein, α-actinin-1 (100 kDa, called ACTN1) binds to the cytosolic C0 domain of GluN1 (residues 841-865) that may play a role in the Ca2+-dependent desensitization of NMDAR channels. Mutations that disrupt NMDAR channel function are linked to Alzheimer's disease, depression, stroke, epilepsy, and schizophrenia. NMR chemical shift assignments are reported here for the C-terminal EF-hand domain of ACTN1 (residues 824-892, called ACTN_EF34) and ACTN_EF34 bound to the GluN1 C0 domain (BMRB numbers 52385 and 52386, respectively).
Collapse
Affiliation(s)
- Aritra Bej
- Departments of Chemistry, University of California, Davis, CA, 95616, USA
| | - Johannes W Hell
- Departments of Pharmacology, University of California, Davis, CA, 95616, USA
| | - James B Ames
- Departments of Chemistry, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
23
|
Ullman EZ, Perszyk RE, Paladugu S, Fritzemeier RG, Akins NS, Jacobs L, Liotta DC, Traynelis SF. Mechanisms of Action Underlying Conductance-Modifying Positive Allosteric Modulators of the NMDA Receptor. Mol Pharmacol 2024; 106:334-353. [PMID: 39443157 PMCID: PMC11585258 DOI: 10.1124/molpharm.124.001019] [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: 08/23/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ionotropic glutamate receptors that mediate a slow, Ca2+-permeable component of excitatory neurotransmission. Modulation of NMDAR function has the potential for disease modification as NMDAR dysfunction has been implicated in neurodevelopment, neuropsychiatric, neurologic, and neurodegenerative disorders. We recently described the thieno[2,3-day]pyrimidin-4-one (EU1622) class of positive allosteric modulators, including several potent and efficacious analogs. Here we have used electrophysiological recordings from Xenopus oocytes, human embryonic kidney cells, and cultured cerebellar and cortical neurons to determine the mechanisms of action of a representative member of this class of modulator. EU1622-240 enhances current response to saturating agonist (doubling response amplitude at 0.2-0.5 μM), slows the deactivation time course following rapid removal of glutamate, increases open probability, enhances coagonist potency, and reduces single-channel conductance. We also show that EU1622-240 facilitates NMDAR activation when only glutamate or glycine is bound. EU1622-240-bound NMDARs channels activated by a single agonist (glutamate or glycine) open to a unique conductance level with different pore properties and Mg2+ sensitivity, in contrast to channels arising from activation of NMDARs with both coagonists bound. These data demonstrate that previously hypothesized distinct gating steps can be controlled by glutamate and glycine binding and shows that the 1622-series modulators enable glutamate- or glycine-bound NMDARs to generate open conformations with different pore properties. The properties of this class of allosteric modulators present intriguing therapeutic opportunities for the modulation of circuit function. SIGNIFICANCE STATEMENT: NMDA receptors are expressed throughout the central nervous system and are permeable to calcium. EU1622-240 increases open probability and agonist potency while reducing single-channel conductance and prolonging the deactivation time course. EU1622-240 allows NMDA receptor activation by the binding of one coagonist (glycine or glutamate), which produces channels with distinct properties. Evaluation of this modulator provides insight into gating mechanisms and may lead to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Elijah Z Ullman
- Departments of Pharmacology and Chemical Biology, Emory School of Medicine (E.Z.U., R.E.P., S.F.T.) and Chemistry (S.P., R.G.F., N.S.A., L.J., D.C.L.), Emory University, Atlanta, Georgia
| | - Riley E Perszyk
- Departments of Pharmacology and Chemical Biology, Emory School of Medicine (E.Z.U., R.E.P., S.F.T.) and Chemistry (S.P., R.G.F., N.S.A., L.J., D.C.L.), Emory University, Atlanta, Georgia
| | - Srinu Paladugu
- Departments of Pharmacology and Chemical Biology, Emory School of Medicine (E.Z.U., R.E.P., S.F.T.) and Chemistry (S.P., R.G.F., N.S.A., L.J., D.C.L.), Emory University, Atlanta, Georgia
| | - Russell G Fritzemeier
- Departments of Pharmacology and Chemical Biology, Emory School of Medicine (E.Z.U., R.E.P., S.F.T.) and Chemistry (S.P., R.G.F., N.S.A., L.J., D.C.L.), Emory University, Atlanta, Georgia
| | - Nicholas S Akins
- Departments of Pharmacology and Chemical Biology, Emory School of Medicine (E.Z.U., R.E.P., S.F.T.) and Chemistry (S.P., R.G.F., N.S.A., L.J., D.C.L.), Emory University, Atlanta, Georgia
| | - Leon Jacobs
- Departments of Pharmacology and Chemical Biology, Emory School of Medicine (E.Z.U., R.E.P., S.F.T.) and Chemistry (S.P., R.G.F., N.S.A., L.J., D.C.L.), Emory University, Atlanta, Georgia
| | - Dennis C Liotta
- Departments of Pharmacology and Chemical Biology, Emory School of Medicine (E.Z.U., R.E.P., S.F.T.) and Chemistry (S.P., R.G.F., N.S.A., L.J., D.C.L.), Emory University, Atlanta, Georgia
| | - Stephen F Traynelis
- Departments of Pharmacology and Chemical Biology, Emory School of Medicine (E.Z.U., R.E.P., S.F.T.) and Chemistry (S.P., R.G.F., N.S.A., L.J., D.C.L.), Emory University, Atlanta, Georgia
| |
Collapse
|
24
|
Zhang XL, Li YX, Berglund N, Burgdorf JS, Donello JE, Moskal JR, Stanton PK. Zelquistinel acts at an extracellular binding domain to modulate intracellular calcium inactivation of N-methyl-d-aspartate receptors. Neuropharmacology 2024; 259:110100. [PMID: 39117105 DOI: 10.1016/j.neuropharm.2024.110100] [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: 06/07/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Stinels are a novel class of N-methyl-d-aspartate glutamate receptor (NMDAR) positive allosteric modulators. We explored mechanism of action and NR2 subtype specificity of the stinel zelquistinel (ZEL) in HEK 293 cells expressing recombinant NMDARs. ZEL potently enhanced NMDAR current at NR2A (EC50 = 9.9 ± 0.5 nM) and NR2C-containing (EC50 = 9.7 ± 0.6 nM) NMDARs, with a larger ceiling enhancement at NR2B-NMDAR (EC50 = 35.0 ± 0.7 nM), while not affecting NR2D-containing NMDARs. In cells expressing NR2A and NR2C-containing NMDARs, ZEL exhibited an inverted-U dose-response relation, with a low concentration enhancement and high concentration suppression of NMDAR currents. Extracellular application of ZEL potentiated NMDAR receptor activity via prolongation of NMDAR currents. Replacing the slow Ca2+ intracellular chelator EGTA with the fast chelator BAPTA blocked ZEL potentiation of NMDARs, suggesting an action on intracellular Ca2+-calmodulin-dependent inactivation (CDI). Consistent with this mechanism of action, removal of the NR1 intracellular C-terminus, or intracellular infusion of a calmodulin blocking peptide, blocked ZEL potentiation of NMDAR current. In contrast, BAPTA did not prevent high-dose suppression of current, indicating this effect has a different mechanism of action. These data indicate ZEL is a novel positive allosteric modulator that binds extracellularly and acts through a unique long-distance mechanism to reduce NMDAR CDI, eliciting enhancement of NMDAR current. The critical role that NMDARs play in long-term, activity-dependent synaptic plasticity, learning, memory and cognition, suggests dysregulation of CDI may contribute to psychiatric disorders such as depression, schizophrenia and others, and that the stinel class of drugs can restore NMDAR-dependent synaptic plasticity by reducing activity-dependent CDI.
Collapse
Affiliation(s)
- Xiao-Lei Zhang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Yong-Xin Li
- AbbVie Inc, 2525 Dupont Drive, Irvine, CA, 92612, USA
| | - Nils Berglund
- Department of Chemistry, Aarhus University, Aarhus, Denmark; Eve BioTek, Warrington, UK
| | | | - John E Donello
- Gate Neurosciences, 1210 Waterway Blvd, Indianapolis, IN, 46202, USA
| | - Joseph R Moskal
- Department of Biomedical Engineering, McCormick School of Engineering and Applied Sciences, Northwestern University, Evanston, IL, 60208, USA
| | - Patric K Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA; Department of Neurology, New York Medical College, Valhalla, NY, 10595, USA.
| |
Collapse
|
25
|
Lüken J, Goerges G, Schreiber JA, Schmidt J, Frehland B, Schepmann D, Seebohm G, Wünsch B. Enantiomerically Pure Indazole Bioisosteres of Ifenprodil and Ro 25-6981 as Negative Allosteric Modulators of NMDA Receptors with the GluN2B Subunit. J Med Chem 2024; 67:19678-19688. [PMID: 39418125 DOI: 10.1021/acs.jmedchem.4c02035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Administration of negative allosteric modulators of GluN2B subunit-containing NMDA receptors such as Ro 25-6981 (1) and ifenprodil (2) results in neuroprotective effects. In this study, the phenol of 1 and 2 was replaced bioisosterically by an indazole to inhibit glucuronidation. The γ- and β-aminoalcohols 10 and 11 were prepared without installing a protective group at the indazole ring using the ketone 6 as a common intermediate. All four stereoisomeric γ- and β-aminoalcohols 10 and 11 were obtained by diastereoselective reduction of ketones 7 and 9 followed by separation of enantiomers. The analogously structured γ-aminoalcohol (1S,2S)-10c (Ro 25-6981 bioisostere) and β-aminoalcohol (1R,2R)-11c (ifenprodil bioisostere) exhibited high GluN2B affinity (Ki = 50 and 66 nM, respectively) and high to moderate inhibitory activity in two-electrode voltage clamp experiments. The indazole bioisosteres 10 and 11 showed higher metabolic stability than 1. In the presence of uridinyldiphosphate activated glucuronic acid, glucuronidation of 10 and 11 was not observed.
Collapse
Affiliation(s)
- Judith Lüken
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, Münster D-48149, Germany
| | - Gunnar Goerges
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster D-48149, Germany
| | - Julian A Schreiber
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, Münster D-48149, Germany
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster D-48149, Germany
| | - Judith Schmidt
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, Münster D-48149, Germany
| | - Bastian Frehland
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, Münster D-48149, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, Münster D-48149, Germany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster D-48149, Germany
- GRK 2515, Chemical biology of ion channels (Chembion), Universität Münster, Corrensstr. 48, Münster D-48149, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, Münster D-48149, Germany
- GRK 2515, Chemical biology of ion channels (Chembion), Universität Münster, Corrensstr. 48, Münster D-48149, Germany
| |
Collapse
|
26
|
Vashisth MK, Hu J, Liu M, Basha SH, Yu C, Huang W. In-Silico discovery of 17alpha-hydroxywithanolide-D as potential neuroprotective allosteric modulator of NMDA receptor targeting Alzheimer's disease. Sci Rep 2024; 14:27908. [PMID: 39537738 PMCID: PMC11560966 DOI: 10.1038/s41598-024-78975-2] [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: 06/02/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder marked by cognitive decline, memory impairment, and behavioral alterations. The N-methyl-D-aspartate (NMDA) receptor has emerged as a promising target for AD pharmacotherapy due to its role in the disease's pathogenesis. This study leverages advanced computational methods to screen 80 active constituents of Withania somnifera (Ashwagandha), a traditional herb known for its neuroprotective effects, against the NMDA receptor, using FDA-approved Ifenprodil as a reference. Our blind virtual screening results demonstrated that all tested compounds could bind to various domains of the NMDA receptor, with binding energies ranging from - 4.1 to -11.9 kcal/mol, compared to Ifenprodil's -7.8 kcal/mol. Binding preference analysis revealed 7 compounds bound to the A-chain, 37 to the B-chain, 7 to the C-chain, and 29 to the D-chain of the receptor. Notable binding was observed predominantly at the Amino Terminal Domain (ATD) core site, some at the ATD-Ligand Binding Domain (LBD) interface, and a few at the Transmembrane Domain (TMD). Particularly, 17alpha-hydroxywithanolide D, with a binding energy of -11.9 kcal/mol, emerged as a prime candidate for further investigation. Molecular dynamics simulations of this compound revealed key interactions, including direct hydrogen bonding with residues ASP165, ARG431, THR433, LYS466, and TYR476 on the D-chain, as well as additional hydrophobic and water-bridging interactions. These simulations highlighted the compound's influence on dynamic conformational states of the GluN1b-GluN2B receptor complex, modulating interactions between GluN1b Lys178 and GluN2B Asn184. Furthermore, the compound affected the distance between LBD heterodimers and the tension within the LBD-M30 linker, demonstrating its potential to modulate NMDA receptor activity. This comprehensive study not only underscores the therapeutic promise of Withania somnifera derivatives for AD but also provides a detailed molecular basis for their efficacy, offering valuable insights for targeted drug development and innovative therapeutic strategies against Alzheimer's disease.
Collapse
Affiliation(s)
- Manoj Kumar Vashisth
- Department of Human Anatomy, School of Basic Medicine Sciences, Southern Medical University, 510515, Guangzhou, P. R. China
| | - Junkai Hu
- Department of Orthopaedics, Affiliated Hospital of Guangdong Medical University, 524001, Zhanjiang, P. R. China
| | - Mingrui Liu
- Department of Human Anatomy, School of Basic Medicine Sciences, Dali University, 671000, Yunnan, China
| | | | - Chen Yu
- Central Laboratory, Affiliated Hospital of Putian University, Putian University, 351100, Putian, China.
| | - Wenhua Huang
- Department of Human Anatomy, School of Basic Medicine Sciences, Southern Medical University, 510515, Guangzhou, P. R. China.
| |
Collapse
|
27
|
Scorrano G, Di Francesco L, Di Ludovico A, Chiarelli F, Matricardi S. Exploring the Landscape of Pre- and Post-Synaptic Pediatric Disorders with Epilepsy: A Narrative Review on Molecular Mechanisms Involved. Int J Mol Sci 2024; 25:11982. [PMID: 39596051 PMCID: PMC11593774 DOI: 10.3390/ijms252211982] [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: 10/07/2024] [Revised: 11/03/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) are a group of conditions affecting brain development, with variable degrees of severity and heterogeneous clinical features. They include intellectual disability (ID), autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), often coexisting with epilepsy, extra-neurological comorbidities, and multisystemic involvement. In recent years, next-generation sequencing (NGS) technologies allowed the identification of several gene pathogenic variants etiologically related to these disorders in a large cohort of affected children. These genes encode proteins involved in synaptic homeostasis, such as SNARE proteins, implicated in calcium-triggered pre-synaptic release of neurotransmitters, or channel subunit proteins, such as post-synaptic ionotropic glutamate receptors involved in the brain's fast excitatory neurotransmission. In this narrative review, we dissected emerged molecular mechanisms related to NDDs and epilepsy due to defects in pre- and post-synaptic transmission. We focused on the most recently discovered SNAREopathies and AMPA-related synaptopathies.
Collapse
Affiliation(s)
- Giovanna Scorrano
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| | - Ludovica Di Francesco
- Department of Neonatology, University of L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy;
| | - Armando Di Ludovico
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| | - Francesco Chiarelli
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| | - Sara Matricardi
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| |
Collapse
|
28
|
Mei T, Chen Y, Gao Y, Zhao H, Lyu X, Lin J, Niu T, Han H, Tong Z. Formaldehyde initiates memory and motor impairments under weightlessness condition. NPJ Microgravity 2024; 10:100. [PMID: 39468074 PMCID: PMC11519943 DOI: 10.1038/s41526-024-00441-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 10/21/2024] [Indexed: 10/30/2024] Open
Abstract
During space flight, prolonged weightlessness stress exerts a range of detrimental impacts on the physiology and psychology of astronauts. These manifestations encompass depressive symptoms, anxiety, and impairments in both short-term memory and motor functions, albeit the precise underlying mechanisms remain elusive. Recent studies have revealed that hindlimb unloading (HU) animal models, which simulate space weightlessness, exhibited a disorder in memory and motor function associated with endogenous formaldehyde (FA) accumulation in the hippocampus and cerebellum, disruption of brain extracellular space (ECS), and blockage of interstitial fluid (ISF) drainage. Notably, the impairment of the blood-brain barrier (BBB) caused by space weightlessness elicits the infiltration of albumin and hemoglobin from the blood vessels into the brain ECS. However, excessive FA has the potential to form cross-links between these two proteins and amyloid-beta (Aβ), thereby obstructing ECS and inducing neuron death. Moreover, FA can inhibit N-methyl-D-aspartate (NMDA) currents by crosslinking NR1 and NR2B subunits, thus impairing memory. Additionally, FA has the ability to modulate the levels of certain microRNAs (miRNAs) such as miRNA-29b, which can affect the expression of aquaporin-4 (AQP4) so as to regulate ECS structure and ISF drainage. Especially, the accumulation of FA may inactivate the ataxia telangiectasia-mutated (ATM) protein kinase by forming cross-linking, a process that is associated with ataxia. Hence, this review presents that weightlessness stress-derived FA may potentially serve as a crucial catalyst in the deterioration of memory and motor abilities in the context of microgravity.
Collapse
Affiliation(s)
- Tianhao Mei
- Beijing Geriatric Hospital, Beijing, China
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Chen
- Beijing Geriatric Hospital, Beijing, China
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yajuan Gao
- Department of Radiology, Peking University Third Hospital, Beijing, China. Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China
- NMPA key Laboratory for Evaluation of Medical Imaging Equipment and Technique, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Hang Zhao
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xingzhou Lyu
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Lin
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tianye Niu
- Shenzhen Bay Laboratory, Shenzhen, China.
- University of Science and Technology of China, Anhui, China.
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China. Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
- NMPA key Laboratory for Evaluation of Medical Imaging Equipment and Technique, Beijing, China.
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China.
| | - Zhiqian Tong
- Beijing Geriatric Hospital, Beijing, China.
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
29
|
Bender PA, Chakraborty S, Durham RJ, Berka V, Carrillo E, Jayaraman V. Bi-directional allosteric pathway in NMDA receptor activation and modulation. Nat Commun 2024; 15:8841. [PMID: 39396999 PMCID: PMC11471786 DOI: 10.1038/s41467-024-53181-w] [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: 04/16/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors are ionotropic glutamate receptors involved in learning and memory. NMDA receptors primarily comprise two GluN1 and two GluN2 subunits. The GluN2 subunit dictates biophysical receptor properties, including the extent of receptor activation and desensitization. GluN2A- and GluN2D-containing receptors represent two functional extremes. To uncover the conformational basis of their functional divergence, we utilize single-molecule fluorescence resonance energy transfer to probe the extracellular domains of these receptor subtypes under resting and ligand-bound conditions. We find that the conformational profile of the GluN2 amino-terminal domain correlates with the disparate functions of GluN2A- and GluN2D-containing receptors. Changes at the pre-transmembrane segments inversely correlate with those observed at the amino-terminal domain, confirming direct allosteric communication between these domains. Additionally, binding of a positive allosteric modulator at the transmembrane domain shifts the conformational profile of the amino-terminal domain towards the active state, revealing a bidirectional allosteric pathway between extracellular and transmembrane domains.
Collapse
Affiliation(s)
- Paula A Bender
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Subhajit Chakraborty
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ryan J Durham
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vladimir Berka
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Elisa Carrillo
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vasanthi Jayaraman
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA.
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
30
|
Cimino M, Feligioni M. The selective disruption of the JNK2/Syntaxin-1A interaction by JGRi1 protects against NMDA-evoked toxicity in SH-SY5Y cells. Neurochem Int 2024; 179:105824. [PMID: 39098765 DOI: 10.1016/j.neuint.2024.105824] [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: 07/03/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
N-methyl-D-aspartate (NMDA) receptors are calcium-permeable ion-channel receptors, specifically activated by glutamate, that permit the activation of specific intracellular calcium-dependent pathways. Aberrant NMDA receptor activation leads to a condition known as excitotoxicity, in which excessive calcium inflow induces apoptotic pathways. To date, memantine is the only NMDA receptor antagonist authorized in clinical practice, hence, a better understanding of the NMDA cascade represents a need to discover novel pharmacological targets. We previously reported non-conventional intracellular signaling triggered by which, upon activation, promotes the interaction between JNK2 and STX1A which enhances the rate of vesicular secretion. We developed a cell-permeable peptide, named JGRi1, able to disrupt such interaction, thus reducing vesicular secretion. In this work, to selectively study the effect of JGRi1 in a much simpler system, we employed neuroblastoma cells, SH-SY5Y. We found that SH-SY5Y cells express the components of the NMDA receptor-JNK2 axis and that the NMDA stimulus increases the rate of vesicle release. Both JGRi1 and memantine protected SH-SY5Y cells from NMDA toxicity, but only JGRi1 reduced the interaction between JNK2 and STX1A. Both drugs successfully reduced NMDA-induced vesicle release, although, unlike memantine, JGRi1 did not prevent calcium influx. NMDA treatment induced JNK2 expression, but not JNK1 or JNK3, which was prevented by both JGRi1 and memantine, suggesting that JNK2 may be specifically involved in the response to NMDA. In conclusion, being JGRi1 able to protect cells against NMDA toxicity by interfering with JNK2/STX1A interaction, it could be considered a novel pharmacological tool to counteract excitotoxicity.
Collapse
Affiliation(s)
- M Cimino
- EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| | - M Feligioni
- EBRI Rita Levi-Montalcini Foundation, Rome, Italy; Department of Neuro-Rehabilitation Sciences, Casa di Cura Igea, Milan, Italy.
| |
Collapse
|
31
|
Wu Y, Zhu Z, Yang J, Wang J, Ji T, Zhu H, Peng W, Chen M, Zhao H. Insights into the terahertz response of L-glutamic acid and its receptor. Analyst 2024; 149:4605-4614. [PMID: 39037577 DOI: 10.1039/d4an00697f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
L-Glutamic acid (L-Glu) is a basic unit of proteins and also serves as an important neurotransmitter in the central nervous system. Its structural properties are critical for biological functions and selective receptor recognition. Although this molecule has been extensively studied, the low frequency vibrational behavior that is closely related to conformational changes and the intermolecular interactions between L-Glu and its receptors are still unclear. In this study, we acquired the fingerprint spectrum of L-Glu by using air plasma terahertz (THz) time-domain spectroscopy in the 0.5-18 THz range. The low frequency vibrational characteristics of L-Glu were investigated through density functional theory (DFT) calculations. The THz responses of the ligand binding domain of the NMDAR-L-Glu complex were studied by the ONIOM method, with a focus on discussing the normal modes and interactions of ligand L-Glu and water molecules. The results illustrate that THz spectroscopy exhibits a sensitive response to the influence of L-Glu on the structure of the NMDAR. The water molecules in proteins have various strong vibration modes in the THz band, showing specificity, diversity and complexity of vibrational behavior. There is potential for influencing and regulating the structural stability of the NMDAR-L-Glu complex through water molecules.
Collapse
Affiliation(s)
- Yu Wu
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongjie Zhu
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Jinrong Yang
- East China Normal University, Shanghai 200241, China
| | - Jie Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Te Ji
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Huachun Zhu
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Weiwei Peng
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Min Chen
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Hongwei Zhao
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| |
Collapse
|
32
|
Rathing F, Schepmann D, Wünsch B. Quinolone bioisosteres of phenolic GluN2B-selective NMDA receptor antagonists. Arch Pharm (Weinheim) 2024; 357:e2400279. [PMID: 38889396 DOI: 10.1002/ardp.202400279] [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: 04/15/2024] [Revised: 05/22/2024] [Accepted: 05/26/2024] [Indexed: 06/20/2024]
Abstract
Cyclopenta[g]quinolones of type 4 were designed with the aim to bioisosterically replace the phenol of potent GluN2B ligands such as ifenprodil and Ro 25-6981 by the quinolone system and to restrict the conformational flexibility of the aminopropanol substructure in a cyclopentane system. The designed ligands were synthesized in an eight-step sequence starting with terephthalaldehyde (5). Key steps pf the synthesis were the intramolecular Friedel-Crafts acylation of propionic acids 10 to yield the cyclopenta[g]quinolinediones 11 and the Mannich reaction of diketone 11a followed by conjugate addition at the α,β-unsaturated ketone 12a. Although the quinolones 13a, 15a, and 16a contain an H-bond donor group (secondary lactam) as ifenprodil and Ro 25-6981, they show only moderate GluN2B affinity (Ki > 410 nM). However, the introduction of lipophilic substituents at the quinolone N-atom resulted in more than 10-fold increased GluN2B affinity of the benzyl and benzyloxymethyl derivatives cis-13c (Ko = 36 nM) and 13e (Ko = 27 nM). All compounds are selective over the phencyclidine (PCP) binding site of the N-methyl-D-aspartate (NMDA) receptor. The benzyl derivative 13c showed six- and threefold selectivity over σ1 and σ2 receptors, respectively.
Collapse
Affiliation(s)
- Friederike Rathing
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Münster, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Münster, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Münster, Germany
- GRK 2515, Chemical biology of ion channels (Chembion), Universität Münster, Münster, Germany
| |
Collapse
|
33
|
Kolić D, Kovarik Z. N-methyl-d-aspartate receptors: Structure, function, and role in organophosphorus compound poisoning. Biofactors 2024; 50:868-884. [PMID: 38415801 DOI: 10.1002/biof.2048] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024]
Abstract
Acute organophosphorus compound (OP) poisoning induces symptoms of the cholinergic crises with the occurrence of severe epileptic seizures. Seizures are induced by hyperstimulation of the cholinergic system, but are enhanced by hyperactivation of the glutamatergic system. Overstimulation of muscarinic cholinergic receptors by the elevated acetylcholine causes glutamatergic hyperexcitation and an increased influx of Ca2+ into neurons through a type of ionotropic glutamate receptors, N-methyl-d-aspartate (NMDA) receptors (NMDAR). These excitotoxic signaling processes generate reactive oxygen species, oxidative stress, and activation of the neuroinflammatory response, which can lead to recurrent epileptic seizures, neuronal cell death, and long-term neurological damage. In this review, we illustrate the NMDAR structure, complexity of subunit composition, and the various receptor properties that change accordingly. Although NMDARs are in normal physiological conditions important for controlling synaptic plasticity and mediating learning and memory functions, we elaborate the detrimental role NMDARs play in neurotoxicity of OPs and focus on the central role NMDAR inhibition plays in suppressing neurotoxicity and modulating the inflammatory response. The limited efficacy of current medical therapies for OP poisoning concerning the development of pharmacoresistance and mitigating proinflammatory response highlights the importance of NMDAR inhibitors in preventing neurotoxic processes and points to new avenues for exploring therapeutics for OP poisoning.
Collapse
Affiliation(s)
- Dora Kolić
- Division of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Zrinka Kovarik
- Division of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
- Department of Chemistry, Faculty of Science, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
34
|
Bleier J, Furtado de Mendonca PR, Habrian CH, Stanley C, Vyklicky V, Isacoff EY. Subtype-specific conformational landscape of NMDA receptor gating. Cell Rep 2024; 43:114634. [PMID: 39154344 PMCID: PMC11446236 DOI: 10.1016/j.celrep.2024.114634] [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: 05/09/2024] [Revised: 06/22/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024] Open
Abstract
N-methyl-D-aspartate receptors are ionotropic glutamate receptors that mediate synaptic transmission and plasticity. Variable GluN2 subunits in diheterotetrameric receptors with identical GluN1 subunits set very different functional properties. To understand this diversity, we use single-molecule fluorescence resonance energy transfer (smFRET) to measure the conformations of the ligand binding domain and modulatory amino-terminal domain of the common GluN1 subunit in receptors with different GluN2 subunits. Our results demonstrate a strong influence of the GluN2 subunits on GluN1 rearrangements, both in non-agonized and partially agonized activation intermediates, which have been elusive to structural analysis, and in the fully liganded state. Chimeric analysis reveals structural determinants that contribute to these subtype differences. Our study provides a framework for understanding the conformational landscape that supports highly divergent levels of activity, desensitization, and agonist potency in receptors with different GluN2s and could open avenues for the development of subtype-specific modulators.
Collapse
Affiliation(s)
- Julia Bleier
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Chris H Habrian
- Biophysics Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Cherise Stanley
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Vojtech Vyklicky
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ehud Y Isacoff
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Weill Neurohub, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biology & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
35
|
Barragan EV, Anisimova M, Vijayakumar V, Coblentz A, Park DK, Salaka RJ, Nisan AFK, Petshow S, Dore K, Zito K, Gray JA. d-Serine Inhibits Non-ionotropic NMDA Receptor Signaling. J Neurosci 2024; 44:e0140242024. [PMID: 38942470 PMCID: PMC11308331 DOI: 10.1523/jneurosci.0140-24.2024] [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: 01/17/2024] [Revised: 05/24/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024] Open
Abstract
NMDA-type glutamate receptors (NMDARs) are widely recognized as master regulators of synaptic plasticity, most notably for driving long-term changes in synapse size and strength that support learning. NMDARs are unique among neurotransmitter receptors in that they require binding of both neurotransmitter (glutamate) and co-agonist (e.g., d-serine) to open the receptor channel, which leads to the influx of calcium ions that drive synaptic plasticity. Over the past decade, evidence has accumulated that NMDARs also support synaptic plasticity via ion flux-independent (non-ionotropic) signaling upon the binding of glutamate in the absence of co-agonist, although conflicting results have led to significant controversy. Here, we hypothesized that a major source of contradictory results might be attributed to variable occupancy of the co-agonist binding site under different experimental conditions. To test this hypothesis, we manipulated co-agonist availability in acute hippocampal slices from mice of both sexes. We found that enzymatic scavenging of endogenous co-agonists enhanced the magnitude of long-term depression (LTD) induced by non-ionotropic NMDAR signaling in the presence of the NMDAR pore blocker MK801. Conversely, a saturating concentration of d-serine completely inhibited LTD and spine shrinkage induced by glutamate binding in the presence of MK801 or Mg2+ Using a Förster resonance energy transfer (FRET)-based assay in cultured neurons, we further found that d-serine completely blocked NMDA-induced conformational movements of the GluN1 cytoplasmic domains in the presence of MK801. Our results support a model in which d-serine availability serves to modulate NMDAR signaling and synaptic plasticity even when the NMDAR is blocked by magnesium.
Collapse
Affiliation(s)
- Eden V Barragan
- Center for Neuroscience, University of California, Davis, California 95618
| | - Margarita Anisimova
- Center for Neuroscience, University of California, Davis, California 95618
- Departments of Neurobiology, Physiology and Behavior, University of California, Davis, California 95618
| | - Vishnu Vijayakumar
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California at San Diego, San Diego, California 92093
| | - Azariah Coblentz
- Center for Neuroscience, University of California, Davis, California 95618
- Departments of Neurobiology, Physiology and Behavior, University of California, Davis, California 95618
| | - Deborah K Park
- Center for Neuroscience, University of California, Davis, California 95618
- Departments of Neurobiology, Physiology and Behavior, University of California, Davis, California 95618
| | - Raghava Jagadeesh Salaka
- Center for Neuroscience, University of California, Davis, California 95618
- Neurology, University of California, Davis, California 95618
| | - Atheer F K Nisan
- Center for Neuroscience, University of California, Davis, California 95618
| | - Samuel Petshow
- Center for Neuroscience, University of California, Davis, California 95618
- Departments of Neurobiology, Physiology and Behavior, University of California, Davis, California 95618
| | - Kim Dore
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California at San Diego, San Diego, California 92093
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, California 95618
- Departments of Neurobiology, Physiology and Behavior, University of California, Davis, California 95618
| | - John A Gray
- Center for Neuroscience, University of California, Davis, California 95618
- Neurology, University of California, Davis, California 95618
- Psychiatry and Behavioral Sciences, University of California, Davis, California 95618
| |
Collapse
|
36
|
Haskel MVL, da Silva Correa V, Queiroz R, Bonini JS, da Silva WC. On the participation of glycine receptors in the reconsolidation of spatial long-term memory in male rats. Behav Brain Res 2024; 471:115086. [PMID: 38825024 DOI: 10.1016/j.bbr.2024.115086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/14/2024] [Accepted: 05/31/2024] [Indexed: 06/04/2024]
Abstract
The effects of intra-hippocampal manipulation of glycine receptors on the reconsolidation of recent and late long-term spatial memory were evaluated and assessed in the Morris water maze. The results obtained from the intra-hippocampal infusion of glycine and taurine demonstrated that taurine at a 100 nmol/side dose impaired the reconsolidation of recent and late long-term spatial memory. In comparison, at a dose of 10 nmol/side, it only affected the reconsolidation of late long-term spatial memory, reinforcing that there are differences between molecular mechanisms underlying recent and late long-term memory reconsolidation. On the other hand, glycine impaired the reconsolidation of early and late spatial memory when infused at a dose of 10 nmol/side, but not at a dose of 100 nmol/side, unless it is co-infused with an allosteric site antagonist of the NMDA receptor. Altogether these results show that glycine acting in situ in the hippocampal CA1 region exerts a pharmacological effect on U-curve, which can be explained by its concomitant action on its ionotropic receptor GlyR and on its NMDA receptor co-agonist site.
Collapse
MESH Headings
- Animals
- Receptors, Glycine/metabolism
- Receptors, Glycine/drug effects
- Male
- Glycine/pharmacology
- Rats
- Spatial Memory/drug effects
- Spatial Memory/physiology
- Memory, Long-Term/drug effects
- Memory, Long-Term/physiology
- Rats, Wistar
- Taurine/pharmacology
- Taurine/administration & dosage
- Hippocampus/drug effects
- Hippocampus/metabolism
- Memory Consolidation/drug effects
- Memory Consolidation/physiology
- Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, N-Methyl-D-Aspartate/drug effects
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/metabolism
- CA1 Region, Hippocampal/physiology
- Maze Learning/drug effects
- Maze Learning/physiology
Collapse
Affiliation(s)
- Maria Vaitsa Loch Haskel
- Program in Physiology, Federal University of Rio Grande do Sul, Porto Alegre 90050-170, Brazil; Laboratory of Neuropsychopharmacology, Department of Pharmacy, State University of Centre-West of Paraná, Guarapuava 85040-167, Brazil
| | - Vinicius da Silva Correa
- Laboratory of Neuropsychopharmacology, Department of Pharmacy, State University of Centre-West of Paraná, Guarapuava 85040-167, Brazil
| | - Ruliam Queiroz
- Program in Physiology, Federal University of Rio Grande do Sul, Porto Alegre 90050-170, Brazil; Laboratory of Neuropsychopharmacology, Department of Pharmacy, State University of Centre-West of Paraná, Guarapuava 85040-167, Brazil
| | - Juliana Sartori Bonini
- Laboratory of Neuropsychopharmacology, Department of Pharmacy, State University of Centre-West of Paraná, Guarapuava 85040-167, Brazil
| | - Weber Claudio da Silva
- Program in Physiology, Federal University of Rio Grande do Sul, Porto Alegre 90050-170, Brazil; Laboratory of Neuropsychopharmacology, Department of Pharmacy, State University of Centre-West of Paraná, Guarapuava 85040-167, Brazil.
| |
Collapse
|
37
|
Chou TH, Epstein M, Fritzemeier RG, Akins NS, Paladugu S, Ullman EZ, Liotta DC, Traynelis SF, Furukawa H. Molecular mechanism of ligand gating and opening of NMDA receptor. Nature 2024; 632:209-217. [PMID: 39085540 PMCID: PMC11376105 DOI: 10.1038/s41586-024-07742-0] [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: 02/19/2024] [Accepted: 06/20/2024] [Indexed: 08/02/2024]
Abstract
Glutamate transmission and activation of ionotropic glutamate receptors are the fundamental means by which neurons control their excitability and neuroplasticity1. The N-methyl-D-aspartate receptor (NMDAR) is unique among all ligand-gated channels, requiring two ligands-glutamate and glycine-for activation. These receptors function as heterotetrameric ion channels, with the channel opening dependent on the simultaneous binding of glycine and glutamate to the extracellular ligand-binding domains (LBDs) of the GluN1 and GluN2 subunits, respectively2,3. The exact molecular mechanism for channel gating by the two ligands has been unclear, particularly without structures representing the open channel and apo states. Here we show that the channel gate opening requires tension in the linker connecting the LBD and transmembrane domain (TMD) and rotation of the extracellular domain relative to the TMD. Using electron cryomicroscopy, we captured the structure of the GluN1-GluN2B (GluN1-2B) NMDAR in its open state bound to a positive allosteric modulator. This process rotates and bends the pore-forming helices in GluN1 and GluN2B, altering the symmetry of the TMD channel from pseudofourfold to twofold. Structures of GluN1-2B NMDAR in apo and single-liganded states showed that binding of either glycine or glutamate alone leads to distinct GluN1-2B dimer arrangements but insufficient tension in the LBD-TMD linker for channel opening. This mechanistic framework identifies a key determinant for channel gating and a potential pharmacological strategy for modulating NMDAR activity.
Collapse
Affiliation(s)
- Tsung-Han Chou
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Max Epstein
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | | | - Srinu Paladugu
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Elijah Z Ullman
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
38
|
Candelas Serra M, Kuchtiak V, Kubik-Zahorodna A, Kysilov B, Fili K, Hrcka Krausova B, Abramova V, Dobrovolski M, Harant K, Bozikova P, Cerny J, Prochazka J, Kasparek P, Sedlacek R, Balik A, Smejkalova T, Vyklicky L. Characterization of Mice Carrying a Neurodevelopmental Disease-Associated GluN2B(L825V) Variant. J Neurosci 2024; 44:e2291232024. [PMID: 38926089 PMCID: PMC11293445 DOI: 10.1523/jneurosci.2291-23.2024] [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: 12/08/2023] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
N-Methyl-d-aspartate receptors (NMDARs), encoded by GRIN genes, are ionotropic glutamate receptors playing a critical role in synaptic transmission, plasticity, and synapse development. Genome sequence analyses have identified variants in GRIN genes in patients with neurodevelopmental disorders, but the underlying disease mechanisms are not well understood. Here, we have created and evaluated a transgenic mouse line carrying a missense variant Grin2bL825V , corresponding to a de novo GRIN2B variant encoding GluN2B(L825V) found in a patient with intellectual disability (ID) and autism spectrum disorder (ASD). We used HEK293T cells expressing recombinant receptors and primary hippocampal neurons prepared from heterozygous Grin2bL825V/+ (L825V/+) and wild-type (WT) Grin2b+/+ (+/+) male and female mice to assess the functional impact of the variant. Whole-cell NMDAR currents were reduced in neurons from L825V/+ compared with +/+ mice. The peak amplitude of NMDAR-mediated evoked excitatory postsynaptic currents (NMDAR-eEPSCs) was unchanged, but NMDAR-eEPSCs in L825V/+ neurons had faster deactivation compared with +/+ neurons and were less sensitive to a GluN2B-selective antagonist ifenprodil. Together, these results suggest a decreased functional contribution of GluN2B subunits to synaptic NMDAR currents in hippocampal neurons from L825V/+ mice. The analysis of the GluN2B(L825V) subunit surface expression and synaptic localization revealed no differences compared with WT GluN2B. Behavioral testing of mice of both sexes demonstrated hypoactivity, anxiety, and impaired sensorimotor gating in the L825V/+ strain, particularly affecting males, as well as cognitive symptoms. The heterozygous L825V/+ mouse offers a clinically relevant model of GRIN2B-related ID/ASD, and our results suggest synaptic-level functional changes that may contribute to neurodevelopmental pathology.
Collapse
Affiliation(s)
- Miriam Candelas Serra
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Viktor Kuchtiak
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
- Faculty of Science, Charles University, Prague 12800, Czech Republic
| | - Agnieszka Kubik-Zahorodna
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec 25050, Czech Republic
| | - Bohdan Kysilov
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Klevinda Fili
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
- Third Faculty of Medicine, Charles University, Prague 10000, Czech Republic
| | | | - Vera Abramova
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
- Third Faculty of Medicine, Charles University, Prague 10000, Czech Republic
| | - Mark Dobrovolski
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
- Third Faculty of Medicine, Charles University, Prague 10000, Czech Republic
| | - Karel Harant
- Proteomics Core Facility, Faculty of Science, Charles University, Biocev, Vestec 25050, Czech Republic
| | - Paulina Bozikova
- Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25050, Czech Republic
| | - Jiri Cerny
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec 25050, Czech Republic
| | - Petr Kasparek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec 25050, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec 25050, Czech Republic
| | - Ales Balik
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Tereza Smejkalova
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Ladislav Vyklicky
- Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| |
Collapse
|
39
|
Li H, Rajani V, Sengar AS, Salter MW. Src dependency of the regulation of LTP by alternative splicing of GRIN1 exon 5. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230236. [PMID: 38853562 PMCID: PMC11343231 DOI: 10.1098/rstb.2023.0236] [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: 10/31/2023] [Revised: 01/31/2024] [Accepted: 02/11/2024] [Indexed: 06/11/2024] Open
Abstract
Alternative splicing of Grin1 exon 5 regulates induction of long-term potentiation (LTP) at Schaffer collateral-CA1 synapses: LTP in mice lacking the GluN1 exon 5-encoded N1 cassette (GluN1a mice) is significantly increased compared with that in mice compulsorily expressing this exon (GluN1b mice). The mechanism underlying this difference is unknown. Here, we report that blocking the non-receptor tyrosine kinase Src prevents induction of LTP in GluN1a mice but not in GluN1b. We find that activating Src enhances pharmacologically isolated synaptic N-methyl-d-aspartate receptor (NMDAR) currents in GluN1a mice but not in GluN1b. Moreover, we observe that Src activation increases the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor component of Schaffer collateral-evoked excitatory post-synaptic potentials in GluN1a mice, but this increase is prevented by blocking NMDARs. We conclude that at these synapses, NMDARs in GluN1a mice are subject to upregulation by Src that mediates induction of LTP, whereas NMDARs in GluN1b mice are not regulated by Src, leading to Src-resistance of LTP. Thus, we have uncovered that a key regulatory mechanism for synaptic potentiation is gated by differential splicing of exon 5 of Grin1. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Hongbin Li
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Vishaal Rajani
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Ameet S. Sengar
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Michael W. Salter
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ONM5S 1A8, Canada
| |
Collapse
|
40
|
Seljeset S, Sintsova O, Wang Y, Harb HY, Lynagh T. Constitutive activity of ionotropic glutamate receptors via hydrophobic substitutions in the ligand-binding domain. Structure 2024; 32:966-978.e6. [PMID: 38677289 DOI: 10.1016/j.str.2024.04.001] [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: 08/11/2023] [Revised: 02/11/2024] [Accepted: 04/02/2024] [Indexed: 04/29/2024]
Abstract
Neurotransmitter ligands electrically excite neurons by activating ionotropic glutamate receptor (iGluR) ion channels. Knowledge of the iGluR amino acid residues that dominate ligand-induced activation would enable the prediction of function from sequence. We therefore explored the molecular determinants of activity in rat N-methyl-D-aspartate (NMDA)-type iGluRs (NMDA receptors), complex heteromeric iGluRs comprising two glycine-binding GluN1 and two glutamate-binding GluN2 subunits, using amino acid sequence analysis, mutagenesis, and electrophysiology. We find that a broadly conserved aspartate residue controls both ligand potency and channel activity, to the extent that certain substitutions at this position bypass the need for ligand binding in GluN1 subunits, generating NMDA receptors activated solely by glutamate. Furthermore, we identify a homomeric iGluR from the placozoan Trichoplax adhaerens that has utilized native mutations of this crucial residue to evolve into a leak channel that is inhibited by neurotransmitter binding, pointing to a dominant role of this residue throughout the iGluR superfamily.
Collapse
Affiliation(s)
- Sandra Seljeset
- Michael Sars Centre, University of Bergen, 5008 Bergen, Norway
| | - Oksana Sintsova
- Michael Sars Centre, University of Bergen, 5008 Bergen, Norway
| | - Yuhong Wang
- Michael Sars Centre, University of Bergen, 5008 Bergen, Norway
| | - Hassan Y Harb
- Concept Life Sciences Limited, Frith Knoll Road, Chapel-en-le-Frith, SK23 0PG High Peak, UK
| | - Timothy Lynagh
- Michael Sars Centre, University of Bergen, 5008 Bergen, Norway.
| |
Collapse
|
41
|
Barragan EV, Anisimova M, Vijayakumar V, Coblentz AC, Park DK, Salaka RJ, Nisan AFK, Petshow S, Dore K, Zito K, Gray JA. D-Serine inhibits non-ionotropic NMDA receptor signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596266. [PMID: 38854020 PMCID: PMC11160797 DOI: 10.1101/2024.05.29.596266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
NMDA-type glutamate receptors (NMDARs) are widely recognized as master regulators of synaptic plasticity, most notably for driving long-term changes in synapse size and strength that support learning. NMDARs are unique among neurotransmitter receptors in that they require binding of both neurotransmitter (glutamate) and co-agonist (e.g. d -serine) to open the receptor channel, which leads to the influx of calcium ions that drive synaptic plasticity. Over the past decade, evidence has accumulated that NMDARs also support synaptic plasticity via ion flux-independent (non-ionotropic) signaling upon the binding of glutamate in the absence of co-agonist, although conflicting results have led to significant controversy. Here, we hypothesized that a major source of contradictory results can be attributed to variable occupancy of the co-agonist binding site under different experimental conditions. To test this hypothesis, we manipulated co-agonist availability in acute hippocampal slices from mice of both sexes. We found that enzymatic scavenging of endogenous co-agonists enhanced the magnitude of LTD induced by non-ionotropic NMDAR signaling in the presence of the NMDAR pore blocker, MK801. Conversely, a saturating concentration of d -serine completely inhibited both LTD and spine shrinkage induced by glutamate binding in the presence of MK801. Using a FRET-based assay in cultured neurons, we further found that d -serine completely blocked NMDA-induced conformational movements of the GluN1 cytoplasmic domains in the presence of MK801. Our results support a model in which d -serine inhibits ion flux-independent NMDAR signaling and plasticity, and thus d -serine availability could serve to modulate NMDAR signaling even when the NMDAR is blocked by magnesium. Significance Statement NMDARs are glutamate-gated cation channels that are key regulators of neurodevelopment and synaptic plasticity and unique in their requirement for binding of a co-agonist (e.g. d -serine) in order for the channel to open. NMDARs have been found to drive synaptic plasticity via non-ionotropic (ion flux-independent) signaling upon the binding of glutamate in the absence of co-agonist, though conflicting results have led to controversy. Here, we found that d -serine inhibits non-ionotropic NMDAR-mediated LTD and LTD-associated spine shrinkage. Thus, a major source of the contradictory findings might be attributed to experimental variability in d -serine availability. In addition, the developmental regulation of d -serine levels suggests a role for non-ionotropic NMDAR plasticity during critical periods of plasticity.
Collapse
|
42
|
Kosenkov AM, Maiorov SA, Gaidin SG. Astrocytic NMDA Receptors. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1045-1060. [PMID: 38981700 DOI: 10.1134/s0006297924060063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 07/11/2024]
Abstract
Astrocytic NMDA receptors (NMDARs) are heterotetramers, whose expression and properties are largely determined by their subunit composition. Astrocytic NMDARs are characterized by a low sensitivity to magnesium ions and low calcium conductivity. Their activation plays an important role in the regulation of various intracellular processes, such as gene expression and mitochondrial function. Astrocytic NMDARs are involved in calcium signaling in astrocytes and can act through the ionotropic and metabotropic pathways. Astrocytic NMDARs participate in the interactions of the neuroglia, thus affecting synaptic plasticity. They are also engaged in the astrocyte-vascular interactions and contribute to the regulation of vascular tone. Astrocytic NMDARs are involved in various pathologies, such as ischemia and hyperammonemia, and their blockade prevents negative changes in astrocytes during these diseases.
Collapse
Affiliation(s)
- Artem M Kosenkov
- Pushchino Scientific Center for Biological Research, Institute of Cell Biophysics of the Russian Academy of Sciences, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Sergei A Maiorov
- Pushchino Scientific Center for Biological Research, Institute of Cell Biophysics of the Russian Academy of Sciences, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Sergei G Gaidin
- Pushchino Scientific Center for Biological Research, Institute of Cell Biophysics of the Russian Academy of Sciences, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
43
|
Kellner S, Berlin S. Rescuing tri-heteromeric NMDA receptor function: the potential of pregnenolone-sulfate in loss-of-function GRIN2B variants. Cell Mol Life Sci 2024; 81:235. [PMID: 38795169 PMCID: PMC11127902 DOI: 10.1007/s00018-024-05243-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: 10/09/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/27/2024]
Abstract
N-methyl-D-aspartate receptors (NMDARs emerging from GRIN genes) are tetrameric receptors that form diverse channel compositions in neurons, typically consisting of two GluN1 subunits combined with two GluN2(A-D) subunits. During prenatal stages, the predominant channels are di-heteromers with two GluN1 and two GluN2B subunits due to the high abundance of GluN2B subunits. Postnatally, the expression of GluN2A subunits increases, giving rise to additional subtypes, including GluN2A-containing di-heteromers and tri-heteromers with GluN1, GluN2A, and GluN2B subunits. The latter emerge as the major receptor subtype at mature synapses in the hippocampus. Despite extensive research on purely di-heteromeric receptors containing two identical GRIN variants, the impact of a single variant on the function of other channel forms, notably tri-heteromers, is lagging. In this study, we systematically investigated the effects of two de novo GRIN2B variants (G689C and G689S) in pure, mixed di- and tri-heteromers. Our findings reveal that incorporating a single variant in mixed di-heteromers or tri-heteromers exerts a dominant negative effect on glutamate potency, although 'mixed' channels show improved potency compared to pure variant-containing di-heteromers. We show that a single variant within a receptor complex does not impair the response of all receptor subtypes to the positive allosteric modulator pregnenolone-sulfate (PS), whereas spermine completely fails to potentiate tri-heteromers containing GluN2A and -2B-subunits. We examined PS on primary cultured hippocampal neurons transfected with the variants, and observed a positive impact over current amplitudes and synaptic activity. Together, our study supports previous observations showing that mixed di-heteromers exhibit improved glutamate potency and extend these findings towards the exploration of the effect of Loss-of-Function variants over tri-heteromers. Notably, we provide an initial and crucial demonstration of the beneficial effects of GRIN2B-relevant potentiators on tri-heteromers. Our results underscore the significance of studying how different variants affect distinct receptor subtypes, as these effects cannot be inferred solely from observations made on pure di-heteromers. Overall, this study contributes to ongoing efforts to understand the pathophysiology of GRINopathies and provides insights into potential treatment strategies.
Collapse
Affiliation(s)
- Shai Kellner
- Dept. of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 1 Efron Bat Galim, Haifa, 3525433, Israel
| | - Shai Berlin
- Dept. of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 1 Efron Bat Galim, Haifa, 3525433, Israel.
| |
Collapse
|
44
|
Juliá-Palacios N, Olivella M, Sigatullina Bondarenko M, Ibáñez-Micó S, Muñoz-Cabello B, Alonso-Luengo O, Soto-Insuga V, García-Navas D, Cuesta-Herraiz L, Andreo-Lillo P, Aguilera-Albesa S, Hedrera-Fernández A, González Alguacil E, Sánchez-Carpintero R, Martín Del Valle F, Jiménez González E, Cean Cabrera L, Medina-Rivera I, Perez-Ordoñez M, Colomé R, Lopez L, Engracia Cazorla M, Fornaguera M, Ormazabal A, Alonso-Colmenero I, Illescas KS, Balsells-Mejía S, Mari-Vico R, Duffo Viñas M, Cappuccio G, Terrone G, Romano R, Manti F, Mastrangelo M, Alfonsi C, de Siqueira Barros B, Nizon M, Gjerulfsen CE, Muro VL, Karall D, Zeiner F, Masnada S, Peterlongo I, Oyarzábal A, Santos-Gómez A, Altafaj X, García-Cazorla Á. L-serine treatment in patients with GRIN-related encephalopathy: a phase 2A, non-randomized study. Brain 2024; 147:1653-1666. [PMID: 38380699 DOI: 10.1093/brain/awae041] [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: 12/20/2023] [Accepted: 01/21/2024] [Indexed: 02/22/2024] Open
Abstract
GRIN-related disorders are rare developmental encephalopathies with variable manifestations and limited therapeutic options. Here, we present the first non-randomized, open-label, single-arm trial (NCT04646447) designed to evaluate the tolerability and efficacy of L-serine in children with GRIN genetic variants leading to loss-of-function. In this phase 2A trial, patients aged 2-18 years with GRIN loss-of-function pathogenic variants received L-serine for 52 weeks. Primary end points included safety and efficacy by measuring changes in the Vineland Adaptive Behavior Scales, Bayley Scales, age-appropriate Wechsler Scales, Gross Motor Function-88, Sleep Disturbance Scale for Children, Pediatric Quality of Life Inventory, Child Behavior Checklist and the Caregiver-Teacher Report Form following 12 months of treatment. Secondary outcomes included seizure frequency and intensity reduction and EEG improvement. Assessments were performed 3 months and 1 day before starting treatment and 1, 3, 6 and 12 months after beginning the supplement. Twenty-four participants were enrolled (13 males/11 females, mean age 9.8 years, SD 4.8), 23 of whom completed the study. Patients had GRIN2B, GRIN1 and GRIN2A variants (12, 6 and 5 cases, respectively). Their clinical phenotypes showed 91% had intellectual disability (61% severe), 83% had behavioural problems, 78% had movement disorders and 58% had epilepsy. Based on the Vineland Adaptive Behavior Composite standard scores, nine children were classified as mildly impaired (cut-off score > 55), whereas 14 were assigned to the clinically severe group. An improvement was detected in the Daily Living Skills domain (P = 0035) from the Vineland Scales within the mild group. Expressive (P = 0.005), Personal (P = 0.003), Community (P = 0.009), Interpersonal (P = 0.005) and Fine Motor (P = 0.031) subdomains improved for the whole cohort, although improvement was mostly found in the mild group. The Growth Scale Values in the Cognitive subdomain of the Bayley-III Scale showed a significant improvement in the severe group (P = 0.016), with a mean increase of 21.6 points. L-serine treatment was associated with significant improvement in the median Gross Motor Function-88 total score (P = 0.002) and the mean Pediatric Quality of Life total score (P = 0.00068), regardless of severity. L-serine normalized the EEG pattern in five children and the frequency of seizures in one clinically affected child. One patient discontinued treatment due to irritability and insomnia. The trial provides evidence that L-serine is a safe treatment for children with GRIN loss-of-function variants, having the potential to improve adaptive behaviour, motor function and quality of life, with a better response to the treatment in mild phenotypes.
Collapse
Affiliation(s)
- Natalia Juliá-Palacios
- Neurometabolic Unit and Synaptic Metabolism Lab, Department of Neurology, Hospital Sant Joan de Déu-IRSJD, CIBERER and MetabERN, 08950 Barcelona, Spain
| | - Mireia Olivella
- Bioinformatics and Bioimaging Group. Faculty of Science, Technology and Engineering, University of Vic-Central University of Catalonia, 08500 Vic, Spain
- Institute for Research and Innovation in Life and Health Sciences (IRIS-CC), University of Vic-Central University of Catalonia, 08500 Vic, Spain
| | - Mariya Sigatullina Bondarenko
- Neurometabolic Unit and Synaptic Metabolism Lab, Department of Neurology, Hospital Sant Joan de Déu-IRSJD, CIBERER and MetabERN, 08950 Barcelona, Spain
| | | | - Beatriz Muñoz-Cabello
- Department of Pediatrics, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | - Olga Alonso-Luengo
- Department of Pediatrics, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | | | - Deyanira García-Navas
- Department of Pediatric Neurology, Complejo Hospitalario Universitario de Cáceres, 10003 Cáceres, Spain
| | | | - Patricia Andreo-Lillo
- Neuropediatric Unit, Pediatric Department, University Hospital of Sant Joan d'Alacant, 03550 Sant Joan d'Alacant, Spain
| | - Sergio Aguilera-Albesa
- Paediatric Neurology Unit, Department of Pediatrics, Hospital Universitario de Navarra, 31008, Pamplona, Spain
| | - Antonio Hedrera-Fernández
- Child Neurology Unit, Pediatrics Department, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | | | | | | | | | | | - Ines Medina-Rivera
- Neurometabolic Unit and Synaptic Metabolism Lab, Department of Neurology, Hospital Sant Joan de Déu-IRSJD, CIBERER and MetabERN, 08950 Barcelona, Spain
| | - Marta Perez-Ordoñez
- Child and Adolescent Mental Health Area, Psychiatry and Psychology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Roser Colomé
- Neurometabolic Unit and Synaptic Metabolism Lab, Department of Neurology, Hospital Sant Joan de Déu-IRSJD, CIBERER and MetabERN, 08950 Barcelona, Spain
| | - Laura Lopez
- Department of Rehabilitation, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - María Engracia Cazorla
- Department of Rehabilitation, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Montserrat Fornaguera
- Department of Rehabilitation, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Aida Ormazabal
- Department of Clinical Biochemistry, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- European Reference Network for Hereditary Metabolic Diseases (MetabERN), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Itziar Alonso-Colmenero
- Pediatric Neurology Department, Hospital Sant Joan de Déu, Full Member of ERN EpiCare, Barcelona University, 08950 Barcelona, Spain
| | - Katia Sofía Illescas
- Neurometabolic Unit and Synaptic Metabolism Lab, Department of Neurology, Hospital Sant Joan de Déu-IRSJD, CIBERER and MetabERN, 08950 Barcelona, Spain
| | - Sol Balsells-Mejía
- Department of Research Promotion and Management. Statistical Support, Hospital Sant Joan de Déu (HSJD), 08950 Barcelona, Spain
| | - Rosanna Mari-Vico
- Neurometabolic Unit and Synaptic Metabolism Lab, Department of Neurology, Hospital Sant Joan de Déu-IRSJD, CIBERER and MetabERN, 08950 Barcelona, Spain
| | - Maria Duffo Viñas
- Neurometabolic Unit and Synaptic Metabolism Lab, Department of Neurology, Hospital Sant Joan de Déu-IRSJD, CIBERER and MetabERN, 08950 Barcelona, Spain
- Child and Adolescent Mental Health Area, Psychiatry and Psychology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Gerarda Cappuccio
- Department of Translational Medical Sciences, Università degli Studi di Napoli 'Federico II', 80125 Naples, Italy
- Telethon Institute of Genetics and Medicine, Department of Pediatrics, Pozzuoli, 80131 Naples, Italy
| | - Gaetano Terrone
- Department of Translational Medical Sciences, Università degli Studi di Napoli 'Federico II', 80125 Naples, Italy
| | - Roberta Romano
- Department of Translational Medical Sciences, Università degli Studi di Napoli 'Federico II', 80125 Naples, Italy
| | - Filippo Manti
- Department of Human Neuroscience, University of Rome La Sapienza, 00185 Roma, Lazio, Italy
| | - Mario Mastrangelo
- Department of Women and Child Health and Uroginecological Sciences, Sapienza University of Rome, 00185 Rome, Italy
- Child Neurology and Psychiatry Unit, Department of Neuroscience/Mental Health, Azienda Ospedaliero-Universitaria Policlinico Umberto I, 00161 Rome, Italy
| | - Chiara Alfonsi
- Department of Human Neuroscience, University of Rome La Sapienza, 00185 Roma, Lazio, Italy
| | - Bruna de Siqueira Barros
- Núcleo de Estudos da Saúde do Adolescente, Programa de Pós-Graduação em Ciências Médicas, Universidade do Estado do Rio de Janeiro, Faculdade de Ciência Médicas, 56066 Rio de Janeiro, RJ, Brazil
| | - Mathilde Nizon
- Service de Génétique Médicale, CHU Nantes, 44093 Nantes, France
| | | | - Valeria L Muro
- Pediatric Neurology Unit, Hospital Britanico Buenos Aires, C1280AEB Buenos Aires, Argentina
| | - Daniela Karall
- Clinic for Paediatrics, Division of Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Fiona Zeiner
- Clinic for Paediatrics, Division of Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Silvia Masnada
- Department of Child Neurology, V. Buzzi Children's Hospital, 20125 Milan, Italy
| | - Irene Peterlongo
- Department of Child Neurology, V. Buzzi Children's Hospital, 20125 Milan, Italy
| | - Alfonso Oyarzábal
- Neurometabolic Unit and Synaptic Metabolism Lab, Department of Neurology, Hospital Sant Joan de Déu-IRSJD, CIBERER and MetabERN, 08950 Barcelona, Spain
| | - Ana Santos-Gómez
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | - Xavier Altafaj
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | - Ángeles García-Cazorla
- Neurometabolic Unit and Synaptic Metabolism Lab, Department of Neurology, Hospital Sant Joan de Déu-IRSJD, CIBERER and MetabERN, 08950 Barcelona, Spain
| |
Collapse
|
45
|
Jiang L, Liu N, Zhao F, Huang B, Kang D, Zhan P, Liu X. Discovery of GluN2A subtype-selective N-methyl-d-aspartate (NMDA) receptor ligands. Acta Pharm Sin B 2024; 14:1987-2005. [PMID: 38799621 PMCID: PMC11119548 DOI: 10.1016/j.apsb.2024.01.004] [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: 09/21/2023] [Revised: 12/04/2023] [Accepted: 12/28/2023] [Indexed: 05/29/2024] Open
Abstract
The N-methyl-d-aspartate (NMDA) receptors, which belong to the ionotropic Glutamate receptors, constitute a family of ligand-gated ion channels. Within the various subtypes of NMDA receptors, the GluN1/2A subtype plays a significant role in central nervous system (CNS) disorders. The present article aims to provide a comprehensive review of ligands targeting GluN2A-containing NMDA receptors, encompassing negative allosteric modulators (NAMs), positive allosteric modulators (PAMs) and competitive antagonists. Moreover, the ligands' structure-activity relationships (SARs) and the binding models of representative ligands are also discussed, providing valuable insights for the clinical rational design of effective drugs targeting CNS diseases.
Collapse
Affiliation(s)
| | | | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Boshi Huang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
46
|
Mousavi H, Rimaz M, Zeynizadeh B. Practical Three-Component Regioselective Synthesis of Drug-Like 3-Aryl(or heteroaryl)-5,6-dihydrobenzo[ h]cinnolines as Potential Non-Covalent Multi-Targeting Inhibitors To Combat Neurodegenerative Diseases. ACS Chem Neurosci 2024; 15:1828-1881. [PMID: 38647433 DOI: 10.1021/acschemneuro.4c00055] [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] [Indexed: 04/25/2024] Open
Abstract
Neurodegenerative diseases (NDs) are one of the prominent health challenges facing contemporary society, and many efforts have been made to overcome and (or) control it. In this research paper, we described a practical one-pot two-step three-component reaction between 3,4-dihydronaphthalen-1(2H)-one (1), aryl(or heteroaryl)glyoxal monohydrates (2a-h), and hydrazine monohydrate (NH2NH2•H2O) for the regioselective preparation of some 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnoline derivatives (3a-h). After synthesis and characterization of the mentioned cinnolines (3a-h), the in silico multi-targeting inhibitory properties of these heterocyclic scaffolds have been investigated upon various Homo sapiens-type enzymes, including hMAO-A, hMAO-B, hAChE, hBChE, hBACE-1, hBACE-2, hNQO-1, hNQO-2, hnNOS, hiNOS, hPARP-1, hPARP-2, hLRRK-2(G2019S), hGSK-3β, hp38α MAPK, hJNK-3, hOGA, hNMDA receptor, hnSMase-2, hIDO-1, hCOMT, hLIMK-1, hLIMK-2, hRIPK-1, hUCH-L1, hPARK-7, and hDHODH, which have confirmed their functions and roles in the neurodegenerative diseases (NDs), based on molecular docking studies, and the obtained results were compared with a wide range of approved drugs and well-known (with IC50, EC50, etc.) compounds. In addition, in silico ADMET prediction analysis was performed to examine the prospective drug properties of the synthesized heterocyclic compounds (3a-h). The obtained results from the molecular docking studies and ADMET-related data demonstrated that these series of 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnolines (3a-h), especially hit ones, can really be turned into the potent core of new drugs for the treatment of neurodegenerative diseases (NDs), and/or due to the having some reactionable locations, they are able to have further organic reactions (such as cross-coupling reactions), and expansion of these compounds (for example, with using other types of aryl(or heteroaryl)glyoxal monohydrates) makes a new avenue for designing novel and efficient drugs for this purpose.
Collapse
Affiliation(s)
- Hossein Mousavi
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| | - Mehdi Rimaz
- Department of Chemistry, Payame Noor University, P.O. Box 19395-3697, Tehran 19395-3697, Iran
| | - Behzad Zeynizadeh
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| |
Collapse
|
47
|
Bender PA, Chakraborty S, Durham RJ, Berka V, Carrillo E, Jayaraman V. Bi-directional allosteric pathway in NMDA receptor activation and modulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589813. [PMID: 38659769 PMCID: PMC11042370 DOI: 10.1101/2024.04.16.589813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
N-methyl-D-aspartate (NMDA) receptors are ionotropic glutamate receptors involved in learning and memory. NMDA receptors primarily comprise two GluN1 and two GluN2 subunits. The GluN2 subunit dictates biophysical receptor properties, including the extent of receptor activation and desensitization. GluN2A- and GluN2D-containing receptors represent two functional extremes. To uncover the conformational basis of their functional divergence, we utilized single-molecule fluorescence resonance energy transfer to probe the extracellular domains of these receptor subtypes under resting and ligand-bound conditions. We find that the conformational profile of the GluN2 amino-terminal domain correlates with the disparate functions of GluN2A- and GluN2D-containing receptors. Changes at the pre-transmembrane segments inversely correlate with those observed at the amino-terminal domain, confirming direct allosteric communication between these domains. Additionally, binding of a positive allosteric modulator at the transmembrane domain shifts the conformational profile of the amino-terminal domain towards the active state, revealing a bidirectional allosteric pathway between extracellular and transmembrane domains.
Collapse
|
48
|
Michalski K, Furukawa H. Structure and function of GluN1-3A NMDA receptor excitatory glycine receptor channel. SCIENCE ADVANCES 2024; 10:eadl5952. [PMID: 38598639 PMCID: PMC11006217 DOI: 10.1126/sciadv.adl5952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/05/2024] [Indexed: 04/12/2024]
Abstract
N-methyl-d-aspartate receptors (NMDARs) and other ionotropic glutamate receptors (iGluRs) mediate most of the excitatory signaling in the mammalian brains in response to the neurotransmitter glutamate. Uniquely, NMDARs composed of GluN1 and GluN3 are activated exclusively by glycine, the neurotransmitter conventionally mediating inhibitory signaling when it binds to pentameric glycine receptors. The GluN1-3 NMDARs are vital for regulating neuronal excitability, circuit function, and specific behaviors, yet our understanding of their functional mechanism at the molecular level has remained limited. Here, we present cryo-electron microscopy structures of GluN1-3A NMDARs bound to an antagonist, CNQX, and an agonist, glycine. The structures show a 1-3-1-3 subunit heterotetrameric arrangement and an unprecedented pattern of GluN3A subunit orientation shift between the glycine-bound and CNQX-bound structures. Site-directed disruption of the unique subunit interface in the glycine-bound structure mitigated desensitization. Our study provides a foundation for understanding the distinct structural dynamics of GluN3 that are linked to the unique function of GluN1-3 NMDARs.
Collapse
|
49
|
Nakagawa T, Wang XT, Miguez-Cabello FJ, Bowie D. The open gate of the AMPA receptor forms a Ca 2+ binding site critical in regulating ion transport. Nat Struct Mol Biol 2024; 31:688-700. [PMID: 38409505 PMCID: PMC11536930 DOI: 10.1038/s41594-024-01228-3] [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: 04/08/2023] [Accepted: 01/18/2024] [Indexed: 02/28/2024]
Abstract
Alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionic acid receptors (AMPARs) are cation-selective ion channels that mediate most fast excitatory neurotransmission in the brain. Although their gating mechanism has been studied extensively, understanding how cations traverse the pore has remained elusive. Here we investigated putative ion and water densities in the open pore of Ca2+-permeable AMPARs (rat GRIA2 flip-Q isoform) at 2.3-2.6 Å resolution. We show that the ion permeation pathway attains an extracellular Ca2+ binding site (site-G) when the channel gate moves into the open configuration. Site-G is highly selective for Ca2+ over Na+, favoring the movement of Ca2+ into the selectivity filter of the pore. Seizure-related N619K mutation, adjacent to site-G, promotes channel opening but attenuates Ca2+ binding and thus diminishes Ca2+ permeability. Our work identifies the importance of site-G, which coordinates with the Q/R site of the selectivity filter to ensure the preferential transport of Ca2+ through the channel pore.
Collapse
Affiliation(s)
- Terunaga Nakagawa
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, Nashville, TN, USA.
- Center for Structural Biology, Vanderbilt University, School of Medicine, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN, USA.
| | - Xin-Tong Wang
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | | | - Derek Bowie
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
50
|
Goodell DJ, Whitby FG, Mellem JE, Lei N, Brockie PJ, Maricq AJ, Eckert DM, Hill CP, Madsen DM, Maricq AV. Mechanistic and structural studies reveal NRAP-1-dependent coincident activation of NMDARs. Cell Rep 2024; 43:113694. [PMID: 38265937 PMCID: PMC11531325 DOI: 10.1016/j.celrep.2024.113694] [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: 06/30/2022] [Revised: 11/27/2023] [Accepted: 01/05/2024] [Indexed: 01/26/2024] Open
Abstract
N-methyl-D-aspartate (NMDA)-type ionotropic glutamate receptors have essential roles in neurotransmission and synaptic plasticity. Previously, we identified an evolutionarily conserved protein, NRAP-1, that is required for NMDA receptor (NMDAR) function in C. elegans. Here, we demonstrate that NRAP-1 was sufficient to gate NMDARs and greatly enhanced glutamate-mediated NMDAR gating, thus conferring coincident activation properties to the NMDAR. Intriguingly, vertebrate NMDARs-and chimeric NMDARs where the amino-terminal domain (ATD) of C. elegans NMDARs was replaced by the ATD from vertebrate receptors-were spontaneously active when ectopically expressed in C. elegans neurons. Thus, the ATD is a primary determinant of NRAP-1- and glutamate-mediated gating of NMDARs. We determined the crystal structure of NRAP-1 at 1.9-Å resolution, which revealed two distinct domains positioned around a central low-density lipoprotein receptor class A domain. The NRAP-1 structure, combined with chimeric and mutational analyses, suggests a model where the three NRAP-1 domains work cooperatively to modify the gating of NMDARs.
Collapse
Affiliation(s)
- Dayton J Goodell
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA
| | - Frank G Whitby
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112-5650, USA
| | - Jerry E Mellem
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA
| | - Ning Lei
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA
| | - Penelope J Brockie
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA
| | | | - Debra M Eckert
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112-5650, USA
| | - Christopher P Hill
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112-5650, USA
| | - David M Madsen
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA
| | - Andres V Maricq
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA.
| |
Collapse
|