1
|
Xiong K, Lou S, Lian Z, Wu Y, Kou Z. The GluN3-containing NMDA receptors. Channels (Austin) 2025; 19:2490308. [PMID: 40235311 PMCID: PMC12005412 DOI: 10.1080/19336950.2025.2490308] [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: 10/23/2024] [Revised: 03/03/2025] [Accepted: 03/31/2025] [Indexed: 04/17/2025] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are heterotetrameric ion channels that play crucial roles in brain function. Among all the NMDAR subtypes, GluN1-N3 receptors exhibit unique agonist binding and gating properties. Unlike "conventional" GluN1-N2 receptors, which require both glycine and glutamate for activation, GluN1-N3 receptors are activated solely by glycine. Furthermore, GluN1-N3 receptors display faster desensitization, reduced Ca2+ permeability, and lower sensitivity to Mg2+ blockage compared to GluN1-N2 receptors. Due to these characteristics, GluN1-N3 receptors are thought to play critical roles in eliminating redundant synapses and pruning spines in early stages of brain development. Recent studies have advanced pharmacological tools for specifically targeting GluN1-N3 receptors and provided direct evidence of these glycine-activated excitatory receptors in native brain tissue. The structural basis of GluN1-N3 receptors has also been elucidated through cryo-EM and artificial intelligence. These findings highlight that GluN1-N3 receptors are not only involved in essential brain functions but also present potential targets for drug development.
Collapse
Affiliation(s)
- Kunlong Xiong
- Department of Pulmonary and Critical Care Medicine, Affiliated First Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Shulei Lou
- Institute of Hospital Management, Linyi People’s Hospital, Linyi, Shandong, China
| | - Zuoyu Lian
- Department of General Practice, Cicheng Town Central Health Center, Ningbo, Zhejiang, China
| | - Yunlin Wu
- Hospital Infection Control Section, Affiliated First Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Zengwei Kou
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Kawabata R, Yamamoto S, Kamimura N, Yao I, Yoshikawa K, Koga K. Cuprizone-induced demyelination provokes abnormal intrinsic properties and excitatory synaptic transmission in the male mouse anterior cingulate cortex. Neuropharmacology 2025; 271:110403. [PMID: 40043749 DOI: 10.1016/j.neuropharm.2025.110403] [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: 11/18/2024] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/09/2025]
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system (CNS). Demyelination in the CNS provokes hyperalgesia, negative emotions, and/or cognitive impairment. Cuprizone (CPZ)-induced demyelination is a major demyelinating disease model for rodents. The anterior cingulate cortex (ACC) is a brain region that is responsible for higher brain functions related to MS symptoms. However, little is known whether CPZ exposure induces demyelination in the ACC coincides with changes to intrinsic neuron properties and synaptic transmission. In this study, we first examined if CPZ exposure induces demyelination in the male mouse ACC. CPZ exposure induced demyelination in the ACC and decreased body weight. In addition, demyelination altered intrinsic properties and excitatory synaptic transmission in layer II/III pyramidal neurons from the ACC as indicated by whole-cell patch-clamp in brain slice preparations. CPZ exposure decreased the frequency of action potentials due to increasing rheobase. At the synapse level, CPZ exposure also suppressed evoked excitatory synaptic transmission to the ACC. Finally, CPZ exposure also changed the kinetics of AMPA and NMDA receptors. These results suggest that CPZ exposure induces demyelination in the ACC coinciding with changes in intrinsic properties, action potentials and excitatory synaptic transmission.
Collapse
Affiliation(s)
- Ryo Kawabata
- Biomedical Chemistry Major, Graduate School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan; Department of Neurophysiology, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Shinji Yamamoto
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Moroyama-Machi, Iruma-Gun, Saitama, Japan
| | - Nana Kamimura
- Biomedical Chemistry Major, Graduate School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Ikuko Yao
- Biomedical Chemistry Major, Graduate School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Keisuke Yoshikawa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Moroyama-Machi, Iruma-Gun, Saitama, Japan.
| | - Kohei Koga
- Department of Neurophysiology, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan.
| |
Collapse
|
3
|
Bernard PB, Castano AM, Buonarati OR, Camp CR, Hell JW, Benke TA. Early life seizures chronically disrupt L-type voltage gated calcium channel regulation of mGluR mediated long term depression via interactions with protein phosphatase 2A. Neurobiol Dis 2025; 209:106884. [PMID: 40147739 DOI: 10.1016/j.nbd.2025.106884] [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: 11/18/2024] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025] Open
Abstract
We probed the dependence of metabotropic glutamate receptor dependent long-term depression (mGluR-LTD) on L-type voltage gated calcium channels (LTCCs). In prior work, we found that in a rat model of early life seizures (ELS), exaggerated mGluR-LTD was partly mediated by LTCCs and protein phosphatase 2A (PP2A). Here, we further investigated the interactive role of LTCCs, PP2A, and protein kinase A (PKA) in this same model. PP2Ac is known to bind CaV1.2 and modulate its function; displacement of PP2A (C subunit, or PP2Ac) as well as PKA phosphorylation of CaV1.2 at serine 1928, result in enhanced CaV1.2 function. We found that ELS enhanced LTCC activity. We further found that pharmacological displacement of PP2Ac (but not PP2B/calcineurin) from CaV1.2 enhanced mGluR-LTD in controls. This was occluded by blockade of PP2A or ELS. The LTCC-dihydropyridine agonist BayK 8644 enhanced mGluR-LTD in controls, which was also occluded by ELS. Up-regulation of both intracellular Ca2+ and PKA activity were implicated in ELS enhancement of mGluR-LTD, as LTD was normalized in ELS by depletion of internal calcium stores or blockade of PKA. These results support a dynamic model of mGluR-LTD regulation by LTCCs through PP2Ac binding and phosphorylation by PKA. This regulation is chronically lost after ELS. Together with our prior work, these studies tie hyperactive LTCCs to the chronic ELS behavioral phenotype that includes abnormal working memory, fear conditioning and socialization.
Collapse
Affiliation(s)
- Paul B Bernard
- Departments of Pediatrics, University of Colorado, School of Medicine, United States of America
| | - Anna M Castano
- Departments of Pediatrics, University of Colorado, School of Medicine, United States of America; Pharmacology, University of Colorado, School of Medicine, United States of America
| | - Olivia R Buonarati
- Pharmacology, University of Colorado, School of Medicine, United States of America; Department of Pharmacology, School of Medicine, University of California, Davis, CA, United States of America
| | - Chad R Camp
- Pharmacology, University of Colorado, School of Medicine, United States of America
| | - Johannes W Hell
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, United States of America
| | - Tim A Benke
- Departments of Pediatrics, University of Colorado, School of Medicine, United States of America; Pharmacology, University of Colorado, School of Medicine, United States of America; Neurology, University of Colorado, School of Medicine, United States of America; Otolaryngology, University of Colorado, School of Medicine, United States of America.
| |
Collapse
|
4
|
Rana KMS, Sonoda T, Sato Y, Kondo Y, Ohtsuka S, Kotani T, Ueno D, Tasumi S. De novo transcriptomic analysis to identify candidate genes potentially related to host recognition during infective stage of Caligus fugu (Crustacea: Copepoda). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101433. [PMID: 39914265 DOI: 10.1016/j.cbd.2025.101433] [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: 12/27/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 03/12/2025]
Abstract
Attachment site recognition for infecting a definite host is crucial for obligate ectoparasites. Caligus fugu, a marine copepod parasite primarily found on pufferfishes, has a unique host-/organ-specificity. The infective stage of the species exclusively infects host fins as the attachment site. However, so far, little is known about the mechanism underlying the specificity of C. fugu. In this study, transcriptomic profiles of the infective first copepodid stage (CI) along with the pre- and a post-infective stage, i.e., second nauplii (NII) and second copepodids (CII), respectively, were investigated. The de novo assembled transcriptome of C. fugu showed that a high number of transcripts showed high homology to those of relative species, Caligus rogercresseyi (94.7 %) and Lepeophtheirus salmonis (91.0 %), suggesting that only a small portion of species-specific genes contribute to interspecific differences, such as host-seeking. Importantly, no gene was noted from the odorant receptors and gustatory receptors families in C. fugu transcripts, similar to L. salmonis genome. Genes related to chemosensing such as the ionotropic glutamate receptors (iGluR) or ionotropic receptors (IRs), viz., GRIA2, GRIA3, GRID2, GRIK2, GRIK3, IR21a, IR25a, IR40a, and IR93a, likely involved in host-seeking, were highly expressed during CI among the three stages. In addition, inositol 1,4,5-triphosphate receptor-associated 2, another potential candidate gene involved in host-seeking, was significantly upregulated in CI compared with that in NII and maintained at the same level in CII. Our present transcriptomic data should offer a foundation for further investigations on various biological aspects, such as the host-/organ-specificity of sea lice.
Collapse
Affiliation(s)
- K M Shakil Rana
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Kagoshima 890-0065, Japan; Department of Aquaculture, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Takanori Sonoda
- Graduate School of Agriculture, Forestry, and Fisheries, Kagoshima University, Kagoshima, Kagoshima 890-8580, Japan
| | - Yoshiki Sato
- Fisheries Laboratory, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Hamamatsu, Shizuoka 431-0214, Japan
| | - Yusuke Kondo
- Seto Inland Sea Carbon-neutral Research Center, Hiroshima University, 5-8-1 Minato-machi, Takehara, Hiroshima 725-0024, Japan
| | - Susumu Ohtsuka
- Seto Inland Sea Carbon-neutral Research Center, Hiroshima University, 5-8-1 Minato-machi, Takehara, Hiroshima 725-0024, Japan
| | - Tomonari Kotani
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Kagoshima 890-0065, Japan
| | - Daisuke Ueno
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Kagoshima 890-0065, Japan; Graduate School of Agriculture, Saga University, Honjo-cho 1, Saga 840-8502, Japan
| | - Satoshi Tasumi
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Kagoshima 890-0065, Japan.
| |
Collapse
|
5
|
Wang J, Yu X, Song S, Zhang Y, Cao J, Jin C, Bao Z, Wang B, Hu J. Transcriptome analysis reveals the immune responses of leopard coral grouper to nervous necrosis virus infection. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110290. [PMID: 40118231 DOI: 10.1016/j.fsi.2025.110290] [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: 11/07/2024] [Revised: 03/06/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
The leopard coral grouper (Plectropomus leopardus) is a key marine aquaculture species that is vulnerable to nervous necrosis virus infection during its larval stage. This infection often causes viral nervous necrosis disease, resulting in significant losses in aquaculture. The brain tissue serves as a target organ for viral invasion in fish, with vacuolization being the most prominent characteristic observed in susceptible individuals. In this study, a transcriptome analysis was conducted using brain tissues from naturally resistant and susceptible P. leopardus to investigate the defensive mechanisms against nervous necrosis virus infection. Histological observations revealed irregular vacuolization in the brains of fish infected with the nervous necrosis virus. Based on the differentially expressed genes, our study revealed a significant upregulation pathway of neuroactive ligand-receptor interaction, glutamatergic synapse, and GABAergic synapse in the resistant group, while COVID-19 infection pathway and NF-kappa B signaling pathway were primarily downregulated in the susceptible group. These findings suggest that neuroprotective and repair mechanisms may enhance resistance to viral infection while suppressing the immune response, thereby preventing hyperactive damage caused by inflammation. The expression profiles of key differentially expressed genes were validated using qRT-PCR. These genes include those related to the nervous system (grin2b, id4, mpz, sema6bb, dab1a, and nmda2d), cytokine interaction (ccr7), and antigen processing and presentation (hsp90aa1). Fluorescence in situ hybridization (FISH) demonstrated virus co-localization at both the tissue and cellular levels. These findings provide insights into the immune mechanism of leopard coral grouper, aiding in the prevention and treatment of nervous necrosis virus infection.
Collapse
Affiliation(s)
- Jingwen Wang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya, China
| | - Xiaofei Yu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya, China
| | - Siqi Song
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya, China
| | - Yiqian Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya, China
| | - Jinlai Cao
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya, China
| | - Chaofan Jin
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya, China
| | - Zhenmin Bao
- Southern Marine Science and Engineer Guangdong Laboratory, Guangzhou, 511458, China; MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya, China
| | - Bo Wang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya, China.
| | - Jingjie Hu
- Southern Marine Science and Engineer Guangdong Laboratory, Guangzhou, 511458, China; MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya, China.
| |
Collapse
|
6
|
Thoreson WB, Bartol TM, Conoan NH, Diamond JS. The architecture of invaginating rod synapses slows glutamate diffusion and shapes synaptic responses. J Gen Physiol 2025; 157:e202413746. [PMID: 40019452 PMCID: PMC11869902 DOI: 10.1085/jgp.202413746] [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: 12/16/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 03/01/2025] Open
Abstract
Synapses of retinal rod photoreceptors involve deep invaginations occupied by second-order rod bipolar cell (RBP) and horizontal cell (HC) dendrites. Synaptic vesicles are released into this invagination at multiple sites beneath an elongated presynaptic ribbon. To study the impact of this architecture on glutamate diffusion and receptor activity, we reconstructed four rod terminals and their postsynaptic dendrites from serial electron micrographs of the mouse retina. We incorporated these structures into anatomically realistic Monte Carlo simulations of neurotransmitter diffusion and receptor activation. By comparing passive diffusion of glutamate in realistic structures with geometrically simplified models, we found that glutamate exits anatomically realistic synapses 10-fold more slowly than previously predicted. Constraining simulations with physiological data, we modeled activity of EAAT5 glutamate transporters in rods, AMPA receptors on HC dendrites, and metabotropic glutamate receptors (mGluR6) on RBP dendrites. Simulations suggested that ∼3,000 EAAT5 populate rod membranes. While uptake by surrounding glial Müller cells retrieves most glutamate released by rods, binding and uptake by EAAT5 influence RBP kinetics. Glutamate persistence allows mGluR6 on RBP dendrites to integrate the stream of vesicles released by rods in darkness. Glutamate's tortuous diffusional path confers quantal variability, as release from nearby ribbon sites exerts larger effects on RBP and HC receptors than release from more distant sites. Temporal integration supports slower sustained release rates, but additional quantal variability can impede postsynaptic detection of changes in release produced by rod light responses. These results show an example of the profound impact that synaptic architecture can have on postsynaptic responses.
Collapse
Affiliation(s)
- Wallace B. Thoreson
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Thomas M. Bartol
- Computational Neurobiology Laboratory, The Salk Institute, La Jolla, CA, USA
| | - Nicholas H. Conoan
- Electron Microscopy Core, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jeffrey S. Diamond
- Synaptic Physiology Section, Division of Intramural Research, National Institute of Neurological Diseases and Strokes, Bethesda, MD, USA
| |
Collapse
|
7
|
Chang H, Li C, Zhu T, Cai S, Chen J, Zhan F, Zeng L, Fang Y, Ye G, Li J, Su J. GLR3.6 T807I Mutation of Casuarina equisetifolia Is Associated With a Decreased JA Response to Insect Feeding by Lymantria xylina. PLANT, CELL & ENVIRONMENT 2025; 48:3185-3198. [PMID: 39718115 DOI: 10.1111/pce.15347] [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: 05/26/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Lymantria xylina is the most important defoliator, damaging the effective coastal windbreak tree species Casuarina equisetifolia. However, the underlying genetic mechanisms through which C. equisetifolia responds to L. xylina attacks remain unknown. Here, we compared the transcriptional, phytohormone and metabolic differences between susceptible (S) and resistant (R) C. equisetifolia cultivars in response to L. xylina feeding. The main L. xylina-induced resistance in C. equisetifolia was a jasmonate (JA) response and JA synthesis was highly induced by L. xylina feeding at both the transcriptional and metabolic levels, thus promoting flavonoid accumulation. The JA response was highly activated by L. xylina feeding on the R but not in the S cultivar, although the JA signalling pathway was intact in both cultivars. We found a single amino acid mutation in the homologues of glutamate receptor-like protein 3.6 (CeGLR3.6T807I) in the S cultivar. Compared with the GLR3.6 homologues in the R cultivar, phosphorylation of CeGLR3.6T807I was not induced by insect feeding, leading to a decreased JA response in the S cultivar. Collectively, this study provides new insights into the function of CeGLR3.6 in regulating the JA response of C. equisetifolia to L. xylina feeding.
Collapse
Affiliation(s)
- Huan Chang
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province, China
| | - Chengli Li
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province, China
| | - Tengfei Zhu
- National Key Laboratory of Wheat Improvement, College of Life Sciences, Shandong Agricultural University, Taian, Shandong Province, China
| | - Shouping Cai
- Fujian Academy of Forestry Sciences, Fuzhou, Fujian Province, China
| | - Jie Chen
- Fujian Academy of Forestry Sciences, Fuzhou, Fujian Province, China
| | - Fangfang Zhan
- Fujian Academy of Forestry Sciences, Fuzhou, Fujian Province, China
| | - Liqiong Zeng
- Fujian Academy of Forestry Sciences, Fuzhou, Fujian Province, China
| | - Yu Fang
- Institute of Soil Fertilizer, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian Province, China
| | - Gongfu Ye
- Fujian Academy of Forestry Sciences, Fuzhou, Fujian Province, China
| | - Jian Li
- Key Laboratory of Forest Ecosystem Process and Management of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province, China
| | - Jun Su
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province, China
| |
Collapse
|
8
|
Cabral DF, Bigliassi M, Morris TP, Gomes-Osman JR, Fried PJ. Integrating neural substrates, diabetes self-management, and behavior change for tailored lifestyle interventions in type-2 diabetes: A neurobehavioral perspective. Neurosci Biobehav Rev 2025; 172:106103. [PMID: 40081438 DOI: 10.1016/j.neubiorev.2025.106103] [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: 10/17/2024] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Understanding the neural mechanisms that underlie behavior change is critical for improving lifestyle management in type 2 diabetes. Individuals with type 2 diabetes face unique challenges in adopting and maintaining healthy behaviors, which can be influenced by alterations in brain function and plasticity, potentially leading to cognitive impairment. Diabetes self-management education and support (DSMES) programs aim to enhance lifestyle changes and promote better health outcomes while reducing cognitive decline. However, complex neural disruptions in critical regions for cognitive processes like the prefrontal cortex and parietal lobe present significant challenges. Given the likely role of neural mechanisms in behavioral choices, understanding how type 2 diabetes affects these neural substrates is crucial for developing effective interventions. Despite considerable research efforts aimed at testing lifestyle interventions, a critical gap remains in understanding the cognitive and behavioral components of successful diabetes self-management, the neural substrates of those components, and the impact of diabetes on those neural substrates. Bridging this gap necessitates a comprehensive examination integrating neural processes, DSMES strategies, and behavior change models. Thus, this perspective review highlights the urgent need to address the knowledge gaps surrounding the neural correlates of diabetes self-management and the integration of behavioral models into intervention frameworks. We propose a hypothesis-generating question and present preliminary findings comparing neural plasticity, executive functions, and lifestyle behavior among individuals with type 2 diabetes, pre-diabetes, and healthy controls. By elucidating the neural substrates underpinning diabetes and exploring their implications for DSMES and readiness to change, we can pave the way for more effective, personalized approaches to diabetes care.
Collapse
Affiliation(s)
- Danylo F Cabral
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA.
| | - Marcelo Bigliassi
- Department of Teaching and Learning, Florida International University, Miami, FL, USA
| | - Timothy P Morris
- Department of Physical Therapy, Movement, and Rehabilitation Sciences, Northeastern University, Boston, MA, USA
| | - Joyce R Gomes-Osman
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Peter J Fried
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Fritzemeier RG, Akins NS, Arcoria PJ, Paladugu S, Ullman EZ, Allen J, Sheikh R, Nocilla KA, McDaniels ED, Coleman EM, Antonoudiou P, D'Erasmo MP, Bartsch P, Sharma SK, Maguire J, Traynelis SF, Liotta DC. Thienopyrimidinone Derivatives as a GluN2B/C/D Biased, Positive Allosteric Modulator of the N-Methyl-d-Aspartate Receptor. J Med Chem 2025. [PMID: 40254917 DOI: 10.1021/acs.jmedchem.4c02912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Positive allosteric modulators (PAMs) of the N-methyl-d-aspartate receptor (NMDAR) have been proposed as therapeutics in several neuropsychiatric indications, including schizophrenia, depression, cognitive dysfunction, and anxiety. In particular, GluN2D-containing NMDARs are highly expressed in inhibitory interneurons and are a target of interest for drug development. Toward that end, we describe our investigation into the GluN2-selective EU 1622 series of PAMs that enhance receptor efficacy, increase agonist potency, prolong deactivation time course, reduce single channel conductance, and limit calcium influx. Through SAR studies of the amide, aryl, and thiophene side chains, we identified analogues with submicromolar potency that preferentially potentiate GluN2B-, GluN2C-, and GluN2D-containing NMDARs. Elaboration of the thiophene side chain to block metabolism resulted in the discovery of EU 1622-240 (25b) with improved metabolic stability, oral bioavailability, and CNS penetration in rodents. Consequently, we present data with EU 1622-240 showing the promising properties of this series as a biased GluN2 potentiator.
Collapse
Affiliation(s)
| | - Nicholas S Akins
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Paul J Arcoria
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Srinu Paladugu
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Elijah Z Ullman
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - James Allen
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Rehan Sheikh
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Kelsey A Nocilla
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Ellington D McDaniels
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Emanuel M Coleman
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02144, United States
| | - Pantelis Antonoudiou
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02144, United States
| | - Michael P D'Erasmo
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Perry Bartsch
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Savita K Sharma
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Jamie Maguire
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02144, United States
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Dennis C Liotta
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
10
|
Rosa JLO, Brivio P, Rossato DR, Fontoura MB, Souza LEM, Fumagalli F, Calabrese F, Burger ME. Tactile stimulation ameliorates haloperidol-induced movement disturbances in rats by promoting neuromodulation on dopaminergic and glutamatergic systems in nigrostriatal brain area. Prog Neuropsychopharmacol Biol Psychiatry 2025:111376. [PMID: 40258486 DOI: 10.1016/j.pnpbp.2025.111376] [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: 11/29/2024] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025]
Abstract
The antipsychotic haloperidol (HAL) primarily antagonizes dopaminergic type-2 receptors. It is known that antipsychotic treatment are commonly related with extrapyramidal syndrome (EPS), which involves movement disorders such as parkinsonism, akathisia and tardive dyskinesia. Tactile stimulation (TS) has been helpful in animal models of drug abuse and depression, raising our interest in evaluating its possible benefits on extrapyramidal HAL-induced side-effects in male adult rats. Subsequently to a sub-chronic model of EPS, TS reversed orofacial dyskinesia and movement impairments induced by HAL and promoted beneficial modulations on dopaminergic and glutamatergic systems in nigrostriatal brain area. Given these outcomes, it is important to consider that: i) TS can help to control and/or reduce movement disorders consequent to antipsychotic treatment; ii) TS can help to better clarify the imbalanced of dopaminergic system haloperidol promotes. Based on our current and previous studies about the benefits of TS, we hypothesize that the TS may represent an important therapeutic target to treat neuromotor disorders originating in the nigrostriatal system.
Collapse
Affiliation(s)
- J L O Rosa
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - P Brivio
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università deglI Studi di Milano, 20133 Milan, Italy
| | - D R Rossato
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - M B Fontoura
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - L E M Souza
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - F Fumagalli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università deglI Studi di Milano, 20133 Milan, Italy
| | - F Calabrese
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università deglI Studi di Milano, 20133 Milan, Italy
| | - M E Burger
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil; Department of Physiology and Pharmacology (UFSM), Santa Maria, RS, Brazil.
| |
Collapse
|
11
|
Zhang B, Wang Q, Zhang Y, Wang H, Kang J, Zhu Y, Wang B, Feng S. Treatment of Insomnia With Traditional Chinese Medicine Presents a Promising Prospect. Phytother Res 2025. [PMID: 40251853 DOI: 10.1002/ptr.8495] [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: 12/05/2024] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/21/2025]
Abstract
Insomnia, a prevalent sleep disorder, significantly impacts global health. While Western medications provide temporary relief, their risks of dependency and cognitive impairment have spurred the search for safer alternatives. Traditional Chinese Medicine (TCM) offers a promising approach to treating insomnia by focusing on harmonizing the balance of Yin and Yang and the functions of internal organs. This review explores recent research advances in TCM for insomnia treatment, integrating classical theories with modern scientific understanding of key pathological mechanisms, including neurotransmitter regulation (GABA, monoamines), immune-inflammatory responses, the HPA axis, and interactions with the gut microbiota. Growing clinical evidence supports the effectiveness of classical TCM prescriptions and treatments like acupuncture in improving sleep quality, particularly when combined with Western medications to enhance efficacy and reduce dependency. However, TCM also has its limitations. Future research directions should focus on modernizing TCM applications, addressing comorbidities associated with insomnia, exploring the role of gut microbiota, and optimizing medicinal and edible homologous products. By integrating traditional knowledge with cutting-edge technologies, TCM holds great potential for advancing personalized and effective insomnia treatments globally.
Collapse
Affiliation(s)
- Boyi Zhang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qianqian Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou, China
| | - Yuhang Zhang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Hanyu Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jingyu Kang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yandi Zhu
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Baiyan Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou, China
| | - Shuying Feng
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou, China
| |
Collapse
|
12
|
Almohmadi NH, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Abdelaziz AM, Jabir MS, Alexiou A, Papadakis M, Batiha GES. Glutamatergic dysfunction in neurodegenerative diseases focusing on Parkinson's disease: Role of glutamate modulators. Brain Res Bull 2025; 225:111349. [PMID: 40252703 DOI: 10.1016/j.brainresbull.2025.111349] [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: 01/16/2025] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder resulting from the degeneration of dopamenergic neurons in the substantia nigra pars compacta (SNpc). Research has predominantly centered on understanding the dysfunction of dopaminergic neurotransmission in PD. Recently, more studies discussed the potential role of other neurotransmitters in PD neuropathology. One of the most important non-dopaminergic neurotransmitters involved in the pathogenesis of PD is glutamate, which is widely involved in glutamatergic neurotransmission in different brain regions, including SNpc. The development and progression of PD neuropathology and levodopa-induced dyskinesias (LID) are associated with glutamate neurotoxicity. Therefore, this review seeks to explore the possible involvement of glutamatergic signaling in PD development and assess the therapeutic potential of glutamate receptor antagonists in treating the disorder.
Collapse
Affiliation(s)
- Najlaa Hamed Almohmadi
- Clinical Nutrition Department, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq; Jabir ibn Hayyan Medical University Al-Ameer Qu, Po. Box (13), Kufa, Najaf, Iraq.
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University-Arish Branch, Arish 45511, Egypt.
| | - Majid S Jabir
- Department of Applied Science, University of Technology-Iraq, Baghdad, Iraq.
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia; University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Research & Development, Funogen, Athens, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten, Herdecke, Heusnerstrasse 40, Wuppertal 42283, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhur University, Damanhur, AlBeheira 22511, Egypt.
| |
Collapse
|
13
|
Santini R, Fuentes E, Maleeva G, Matera C, Riefolo F, Berrocal JA, Albertazzi L, Gorostiza P, Pujals S. Discotic amphiphilic supramolecular polymers for drug release and cell activation with light. NANOSCALE 2025. [PMID: 40202720 DOI: 10.1039/d4nr02957g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The limited efficacy shown by drug delivery systems so far prompts the development of new molecular approaches for releasing drugs in a controlled and selective manner. Light is a privileged stimulus for delivery because it can be applied in sharp spatiotemporal patterns and is orthogonal to most biological processes. Supramolecular polymers form molecular nanostructures whose robustness, versatility, and responsivity to different stimuli have generated wide interest in materials chemistry. However, their application as drug delivery vehicles has received little attention. We built supramolecular polymers based on discotic amphiphiles that self-assemble in linear nanostructures in water. They can integrate diverse amphiphilic bioligands and release them upon illumination, acutely producing functional effects under physiological conditions. We devised two strategies for drug incorporation into the photoswitchable nanofibers. In the co-assembly strategy, discotic monomers with and without conjugated bioligands were co-assembled in helicoidal supramolecular fibers. In the drug embedding approach, we integrated a potent agonist of muscarinic receptors into the discotic polymer by noncovalent stacking interactions. This ligand can be released on demand with light ex situ and in situ, rapidly activating the target receptor and triggering intracellular responses. These novel discotic supramolecular polymers can be light-driven drug carriers for small, planar, and amphiphilic drugs.
Collapse
Affiliation(s)
- Ramona Santini
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
- CIBER-BBN, ISCIII, Madrid, 28029, Spain
- Doctorate Program in Organic Chemistry, University of Barcelona, Carrer Martí i Franquès, Barcelona, 08028, Spain
| | - Edgar Fuentes
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
| | - Galyna Maleeva
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
- CIBER-BBN, ISCIII, Madrid, 28029, Spain
| | - Carlo Matera
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
- CIBER-BBN, ISCIII, Madrid, 28029, Spain
| | - Fabio Riefolo
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
- CIBER-BBN, ISCIII, Madrid, 28029, Spain
| | - José Augusto Berrocal
- Institute of Chemical Research of Catalonia (ICIQ), Barcelona Institute of Science and Technology (BIST), Av. Països Catalans, 16, Tarragona, E-43007, Spain
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
- CIBER-BBN, ISCIII, Madrid, 28029, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, 08010, Spain.
| | - Silvia Pujals
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia, Barcelona, 08034, Spain.
| |
Collapse
|
14
|
Ba F, Wei J, Feng QY, Yu CY, Song MX, Hu S, Xu GY, Zhang HL, Jiang GQ. GluR2 overexpression in ACC glutamatergic neurons alleviates cancer-induced bone pain in rats. Mol Med 2025; 31:130. [PMID: 40197156 PMCID: PMC11974031 DOI: 10.1186/s10020-025-01183-9] [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: 07/29/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Cancer-induced bone pain (CIBP) is a complex chronic pain with poorly understood mechanisms. The anterior cingulate cortex (ACC) plays a critical role in processing and modulating chronic pain. This study investigates how the GluR2 receptors (calcium impermeable AMPA receptors) in ACC glutamatergic neurons regulate CIBP. METHODS The CIBP models were established by injecting Walker 256 cells into the tibia of SD rats. Paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) were used as indicators of hyperalgesia. The immunofluorescence staining was employed to detect the expression of c-Fos in ACC and identify the subtypes of co-labeled c-Fos+ neurons. Real-time monitoring of calcium activity in ACC glutamatergic neurons was achieved through the fiber photometry. The excitability of glutamatergic neurons in ACC was modulated using chemicalgenetics and optogenetics techniques. The expression of GluR2 at the mRNA and protein level in ACC were assessed using RT-qPCR and Western blotting. RESULTS There were significant reductions in PWT and PWL of CIBP rats after Walker 256 cell injection. The ACC of CIBP rats showed increased c-Fos expression compared to sham rats, with mainly activated c-Fos co-localized with glutamatergic neurons. Optogenetic or chemogenetic activation of ACC glutamatergic neurons led to increased hyperalgesia in sham rats, while suppression of their activity alleviated hyperalgesia in CIBP rats. Calcium activity in ACC glutamatergic neurons of CIBP rats was increased with suprathreshold stimulation of von Frey filament. Notably, surface GluR2 protein and mRNA were reduced in ACC of CIBP rats. Furthermore, overexpression of GluR2 by AAV-CaMKII-GluR2 injection was decreased c-Fos expression in ACC and alleviated hyperalgesia in CIBP rats. CONCLUSIONS These findings suggest that decreased surface GluR2 receptors in ACC glutamatergic neurons contribute to calcium activity and excessive excitability, thereby inducing CIBP in rats. Conversely, GluR2 overexpression in ACC glutamatergic neurons alleviates CIBP in rats. This study provides a new potential therapeutic approach for targeting the GluR2 receptor to alleviate CIBP for cancer patients.
Collapse
Affiliation(s)
- Futing Ba
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, China
| | - Jinrong Wei
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, China
| | - Qi-Yan Feng
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, China
| | - Chen-Yang Yu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, China
| | - Meng-Xue Song
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, China
| | - Shufen Hu
- Laboratory for Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Guang-Yin Xu
- Laboratory for Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Hai-Long Zhang
- Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215123, China.
| | - Guo-Qin Jiang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, China.
| |
Collapse
|
15
|
Holter KM, Klausner MG, Hite MH, Moriarty CT, Barth SH, Pierce BE, Iannucci AN, Sheffler DJ, Cosford NDP, Bimonte-Nelson HA, Raab-Graham KF, Gould RW. 17β-estradiol status alters NMDAR function and antipsychotic-like activity in female rats. Mol Psychiatry 2025:10.1038/s41380-025-02996-0. [PMID: 40185905 DOI: 10.1038/s41380-025-02996-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 03/14/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
Low 17β-estradiol (E2) in females of reproductive age, and marked E2 decline with menopause, contributes to heightened symptom severity in schizophrenia (i.e. cognitive dysfunction) and diminished response to antipsychotic medications. However, the underlying mechanisms are unknown. N-methyl-D-aspartate receptor (NMDAR) hypofunction contributes to the pathophysiology of schizophrenia, yet impact of E2 depletion on NMDAR function is not well characterized. Quantitative electroencephalography (qEEG), specifically gamma power, is a well-established functional readout of cortical activity that is elevated in patients with schizophrenia and is sensitive to alterations in NMDAR function. Using qEEG and touchscreen cognitive assessments, present studies investigated the effects of E2 on NMDAR function by administering MK-801 (NMDAR antagonist) to ovariectomized rats with or without E2 implants (Ovx+E and Ovx, respectively). Ovx rats were more sensitive to MK-801-induced elevations in gamma power and attentional impairments compared to Ovx+E rats. Further investigation revealed these effects were mediated by reduced synaptic GluN2A expression. Consistent with clinical reports, olanzapine (second-generation antipsychotic) was less effective in mitigating MK-801-induced elevations in gamma power in Ovx rats. Lastly, we examined antipsychotic-like activity of a Group II metabotropic glutamate receptor (mGlu2/3) positive allosteric modulator (PAM), SBI-0646535, as a novel therapeutic in E2-deprived conditions. SBI-0646535 reversed MK-801-induced elevations in gamma power regardless of E2 status. Collectively, these studies established a relationship between E2 deprivation and NMDAR function that is in part GluN2A-dependent, supporting the notion that E2 deprivation increases susceptibility to NMDAR hypofunction. This highlights the need to examine age/hormone-specific factors when considering antipsychotic response and designing novel pharmacotherapies.
Collapse
Affiliation(s)
- Kimberly M Holter
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - McKenna G Klausner
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Mary Hunter Hite
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Carson T Moriarty
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Samuel H Barth
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Bethany E Pierce
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Alexandria N Iannucci
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Douglas J Sheffler
- Cancer Molecular Therapeutics Program. NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Nicholas D P Cosford
- Cancer Molecular Therapeutics Program. NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Kimberly F Raab-Graham
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Robert W Gould
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
16
|
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
|
17
|
Corti E, Duarte CB. FMRP Controls Neuronal Architecture and Synaptic Content of NMDA Receptors in Cultured Hippocampal Neurons. J Mol Neurosci 2025; 75:44. [PMID: 40172581 PMCID: PMC11965214 DOI: 10.1007/s12031-025-02325-8] [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: 01/04/2025] [Accepted: 02/20/2025] [Indexed: 04/04/2025]
Abstract
Fragile X syndrome is the most common inherited form of intellectual disability and is caused by the transcriptional silencing of the Fmr1 gene and the lack of fragile X messenger ribonucleoprotein (FMRP). FMRP is an RNA-binding protein that regulates the synthesis of synaptic proteins which are essential for proper brain function. Although circuit hyperexcitability is a hallmark of fragile X syndrome (FXS), the cell-autonomous effects of FMRP deficiency remain poorly understood. In this work, we investigated the functional consequences of the absence of FMRP on neuronal morphology and on ionotropic glutamate receptor surface distribution, using primary cultures of mice hippocampal neurons isolated from wild-type (WT) and Fmr1 knock-out (KO) pups. MAP2 staining of Fmr1 KO neurons showed a decrease in total dendritic length and complexity of the dendritic tree, accompanied by an increase in soma size compared to WT neurons. Moreover, immunolabelling of surface glutamate receptors performed under non-permeabilising conditions showed that Fmr1 KO neurons presented a higher content of synaptic surface GluN2A and a lower content of GluN2B subunits of NMDA receptors, while GluA1 and GluA2 distribution remained unchanged. Finally, multielectrode array data showed that Fmr1 KO neurons presented reduced spontaneous activity compared to control neurons. These data support the hypothesis that at the cellular level, Fmr1 KO hippocampal neurons are less excitable due to altered input processing, driven by structural defects and altered GluN2A expression in the synaptic plasma membrane.
Collapse
Affiliation(s)
- Elisa Corti
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
- Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Carlos B Duarte
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
18
|
Ji RL, Tao YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther 2025; 268:108804. [PMID: 39904401 DOI: 10.1016/j.pharmthera.2025.108804] [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: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Receptors are crucial for converting chemical and environmental signals into cellular responses, making them prime targets in drug discovery, with about 70% of drugs targeting these receptors. Biased signaling, or functional selectivity, has revolutionized drug development by enabling precise modulation of receptor signaling pathways. This concept is more firmly established in G protein-coupled receptor and has now been applied to other receptor types, including ion channels, receptor tyrosine kinases, and nuclear receptors. Advances in structural biology have further refined our understanding of biased signaling. This targeted approach enhances therapeutic efficacy and potentially reduces side effects. Numerous biased drugs have been developed and approved as therapeutics to treat various diseases, demonstrating their significant therapeutic potential. This review provides a comprehensive overview of biased signaling in drug discovery and disease treatment, highlighting recent advancements and exploring the therapeutic potential of these innovative modulators across various diseases.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
19
|
Zhou C, Segura-Covarrubias G, Tajima N. Structural Insights into Kainate Receptor Desensitization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645769. [PMID: 40236080 PMCID: PMC11996427 DOI: 10.1101/2025.03.27.645769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Kainate receptors (KARs), along with AMPA and NMDA receptors, belong to the ionotropic glutamate receptor (iGluR) family and play critical roles in mediating excitatory neurotransmission throughout the central nervous system. KARs also regulate neurotransmitter release and modulate neuronal excitability and plasticity. Receptor desensitization plays a critical role in modulating the strength of synaptic transmission and synaptic plasticity. While KARs share overall structural similarity with AMPA receptors, the desensitized state of KARs differs strikingly from that of other iGluRs. Despite extensive studies on KAR structure and function, a fundamental question remains unanswered: why do KARs require large conformational changes upon desensitization, unlike other iGluRs? To address this, we present cryo-electron microscopy structures of GluK2 with double cysteine mutations in non-desensitized, shallow-desensitized and deep-desensitized conformations. In the shallow-desensitized conformation, two cysteine crosslinks stabilize the receptors in a conformation that resembles the desensitized state of AMPA receptors. However, unlike the tightly closed pore observed in the deep-desensitized KAR and desensitized AMPAR conformations, the channel pore in the shallow-desensitized state remains incompletely closed. Patch-clamp recordings and fluctuation analysis suggest that this state remains ion-permeable, indicating that the lateral rotational movement of KAR ligand-binding domains (LBDs) is critical for complete channel closure and stable desensitization. Together with the different degrees of desensitized conformations, our results define the unique mechanism and conformational dynamics of KAR desensitization. Highlights We present the cryo-EM structure of GluK2 kainate receptors with engineered cysteine crosslinks at the inter-dimer interface, which restricts subunit lateral rotation and attenuates receptor desensitization.The structure of GluK2 double cysteine mutant in complex with the allosteric potentiator BPAM344 and glutamate represents a non-desensitized state, highlighting the critical conformational changes required for ion channel gating.The glutamate-bound GluK2 mutant adopts multiple conformations, representing both shallow- and deep-desensitized states. Electrophysiological recordings indicate that the mutant recovers from desensitization more rapidly, resembling AMPARs. Our structural and functional data suggest that shallow-desensitized KARs remain conductive, implying that the large lateral LBD rotation during KAR desensitization is essential for complete channel closure, distinguishing KARs from other iGluRs.
Collapse
|
20
|
Yasmin F, Marwick KFM, Hunter DW, Nawaz S, Marshall GF, Booker SA, Hardingham GE, Kind PC, Wyllie DJA. Absence of GluN2A in hippocampal CA1 neurons leads to altered dendritic structure and reduced frequency of miniature excitatory synaptic events. Brain Commun 2025; 7:fcaf124. [PMID: 40226380 PMCID: PMC11986202 DOI: 10.1093/braincomms/fcaf124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/04/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
GluN2A is a NMDA receptor subunit postulated as important for learning and memory. In humans, heterozygous loss of function variants in the gene encoding it (GRIN2A) increase the risk of epilepsy, intellectual disability and schizophrenia. Haploinsufficient mouse models show electrophysiological abnormalities and thus to improve and widen understanding of the pathogenesis of GRIN2A-associated disorders in humans, this study aimed to assess the impact of Grin2a absence and haploinsufficiency on core neuronal and synaptic properties in genetically modified rats. Electrophysiological whole-cell current- and voltage-clamp recordings were made from CA1 pyramidal neurons in acute hippocampal slices from wild-type and Grin2a heterozygous (Grin2a+/- ) and homozygous (Grin2a-/- ) knock out rats aged postnatal day 27-34. While reduced levels or absence of GluN2A did not affect neuronal excitability or intrinsic membrane properties in both Grin2a+/- and Grin2a-/- rats, we found a reduced frequency of miniature excitatory post synaptic currents and a reduced density of proximal dendrites suggestive of a reduced number of excitatory synapses. Recordings from CA1 neurons in slices prepared from Grin2a+/- and Grin2a-/- rats revealed there was a reduced ratio of the current mediated by NMDA receptors compared to AMPA receptors, while in Grin2a-/- recordings, there was a slowing of the decay time-constant of the NMDA receptor-mediated excitatory postsynaptic currents. Moreover, neither summation of sub-threshold excitatory postsynaptic potentials nor summation of supra-threshold excitatory postsynaptic potentials to initiate action potential firing in CA1 pyramidal neurons indicated any dependence on GluN2A. We conclude that reduced levels of GluN2A alters the kinetics of NMDA receptor-mediated synaptic events and dendritic structure of CA1 neurons, but do not affect several other core neuronal functions. These relatively subtle changes are consistent with the largely intact neural functioning of the majority of humans carrying GRIN2A loss of function variants. Further research could explore whether the changes in synaptic properties we observed contribute to alterations in higher level circuit dynamics and computation, which may manifest as disorders of cognition and excitability in humans.
Collapse
Affiliation(s)
- Farhana Yasmin
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Katie F M Marwick
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Daniel W Hunter
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Sarfaraz Nawaz
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Grant F Marshall
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4SB, UK
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| |
Collapse
|
21
|
Kumar Mondal A, Carrillo E, Jayaraman V, Twomey EC. Glutamate gating of AMPA-subtype iGluRs at physiological temperatures. Nature 2025:10.1038/s41586-025-08770-0. [PMID: 40140570 DOI: 10.1038/s41586-025-08770-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/11/2025] [Indexed: 03/28/2025]
Abstract
Ionotropic glutamate receptors (iGluRs) are tetrameric ligand-gated ion channels that mediate most excitatory neurotransmission1. iGluRs are gated by glutamate, where on glutamate binding, they open their ion channels to enable cation influx into postsynaptic neurons, initiating signal transduction1,2. The structural mechanics of how glutamate gating occurs in full-length iGluRs is not well understood. Here, using the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid subtype iGluR (AMPAR), we identify the glutamate-gating mechanism. AMPAR activation by glutamate is augmented at physiological temperatures. By preparing AMPARs for cryogenic-electron microscopy at these temperatures, we captured the glutamate-gating mechanism. Activation by glutamate initiates ion channel opening that involves all ion channel helices hinging away from the pore axis in a motif that is conserved across all iGluRs. Desensitization occurs when the local dimer pairs decouple and enables closure of the ion channel below through restoring the channel hinges and refolding the channel gate. Our findings define how glutamate gates iGluRs, provide foundations for therapeutic design and demonstrate how physiological temperatures can alter iGluR function.
Collapse
Affiliation(s)
- Anish Kumar Mondal
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elisa Carrillo
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vasanthi Jayaraman
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, USA.
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Edward C Twomey
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Beckman Center for Cryo-EM at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA.
| |
Collapse
|
22
|
Neuronal receptors at body temperature reveal their gating mechanics. Nature 2025:10.1038/d41586-025-00865-y. [PMID: 40140509 DOI: 10.1038/d41586-025-00865-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
|
23
|
Karsan N, Luiza Bastos A, Goadsby PJ. Glutamate as a Therapeutic Substrate in Migraine. Int J Mol Sci 2025; 26:3023. [PMID: 40243659 PMCID: PMC11988557 DOI: 10.3390/ijms26073023] [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: 02/16/2025] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Recurrent and intense headache is a well appreciated cardinal feature of migraine, a common and incapacitating neurological disorder. Often, there are associated canonical sensory abnormalities, such as light and sound sensitivity, as well as associated nausea. Given this phenotype of disordered sensory processing and, in a third of patients, the phenomenon called aura accompanying migraine attacks, it has been suggested that the pathophysiology of migraine is likely to involve glutamate, the main excitatory neurotransmitter in the central nervous system (CNS). Glutamate plays a role in nociception, central sensitization, and cortical spreading depression (CSD), three processes that are deemed important in migraine biology. With an emphasis on the therapeutic potential of targeting various glutamate receptors in migraine, this review will discuss the currently available literature and emerging findings on the role of targeting glutamatergic pathways for the treatment of migraine. A thorough literature review was carried out on the functions of both metabotropic glutamate receptors (mGluRs), and the ionotropic glutamate receptors (NMDA, AMPA, and kainate) in migraine pathogenesis. The ever-present need for new treatments, the role of glutamate in the migraine aura phenomenon, and the consequences of monogenic migraine mutations on mediating prolonged, complex, or permanent aura are all discussed, culminating in a suggestion that glutamatergic targeting may hold particular promise in the management of migraine aura. There are plausible roles for metabotropic receptors in regulating pain processing in important migraine-related brain structures, like the thalamus and trigeminal nucleus. Similarly, ionotropic receptors contribute to excitatory neurotransmission and neuronal hyperexcitability. Recent studies have shown preclinical and early clinical results for treatments targeting these receptors, but there are still significant issues with treatment response, including drug transport, side effects, and efficacy. With ongoing and emerging discoveries in the field, there is increasing promise of new migraine medications targeting glutamate receptors. For bench to bedside translation in this area, continued study of the molecular basis of migraine, receptor subtypes, and exploration of potential drug delivery methods are needed.
Collapse
Affiliation(s)
- Nazia Karsan
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (N.K.); (A.L.B.)
| | - Alves Luiza Bastos
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (N.K.); (A.L.B.)
| | - Peter J. Goadsby
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (N.K.); (A.L.B.)
- NIHR King’s Clinical Research Facility and SLaM Biomedical Research Centre, King’s College Hospital, London SE5 9RS, UK
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
24
|
Du Y, Luo X, Ye X, Song M, Li Y, Yang S, Huang C, Cui J. Molecular imprinted photoresponse hybrid biocatalyst for ultrasensitive glutamate detection. Int J Biol Macromol 2025; 308:142540. [PMID: 40147642 DOI: 10.1016/j.ijbiomac.2025.142540] [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: 01/09/2025] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
A novel construction strategy for hybrid nanobiocatalyst was developed in this study by combining molecular imprinting technology with Fe3O4 nanoparticles hybrid MOF and enzyme immobilization technology. The impacts of molecular imprinting and the photothermal effect of Fe3O4 were investigated on the catalytic performance of the hybrid nanobiocatalyst. It was demonstrated that Fe3O4 nanoparticles exhibited a photothermal effect when exposed to multi-wavelength light. Leveraging the selective recognition ability of the molecular imprinting technique, the hybrid nanobiocatalyst showed a 15.8 % increase in catalytic activity for glutamate oxidase. Furthermore, under the influence of red light (650 nm), the photothermal effect induced by Fe3O4 accelerated the enzymatic reaction rate, resulting in a 23 % increase in enzyme activity. Benefiting from the specific recognition of the substrate by the Fe3O4 imprinted polymer, the hybrid nanobiocatalyst exhibited the rapid and highly sensitive detection of glutamate. Notably, this work provides an efficient strategy for constructing hybrid nanobiocatalyst with excellent properties.
Collapse
Affiliation(s)
- Yingjie Du
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science and Technology, Tianjin 300457, China; National Key Laboratory for the Development and Utilization of Forest Food Resources, Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Forest Chemistry and Materials International Innovation Highland, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210000, China.
| | - Xiuyan Luo
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xiaohong Ye
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Meijia Song
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yingjia Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shumao Yang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Caoxing Huang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jiandong Cui
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science and Technology, Tianjin 300457, China.
| |
Collapse
|
25
|
Karakuş F, Kuzu B. Mechanistic Analysis of Decabromodiphenyl Ether-Induced Neurotoxicity in Humans Using Network Toxicology and Molecular Docking. Neurotox Res 2025; 43:17. [PMID: 40123016 PMCID: PMC11930881 DOI: 10.1007/s12640-025-00741-7] [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: 01/22/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Commercial decabromodiphenyl ether (c-decaBDE) is a widely used additive flame retardant in textiles and plastics. This formulation predominantly consists of the congener BDE-209, with trace amounts of other brominated diphenyl ether congeners, such as nonabromodiphenyl ether and octabromodiphenyl ether. Recognized as a persistent organic pollutant due to its potential for long-range environmental transport, c-decaBDE poses significant environmental threats and serious human health risks, including endocrine, reproductive, developmental, and neurotoxic effects. The mechanisms underlying its neurotoxicity remain largely undefined. This study investigates the neurotoxic effects of BDE-209 in humans through network toxicology, multi-level bioinformatics approaches, and molecular docking analyses. Prediction results indicate that BDE-209 can cross the blood-brain barrier, entering the central nervous system and inducing neurotoxic effects. A comprehensive analysis has identified 294 potential targets linked to the neurotoxicity induced by BDE-209. Gene-gene interaction and pathway enrichment analyses revealed significant associations related to cellular responses to chemical stress and synaptic transmission. Further investigation of protein-protein interactions, combined with centrality analysis, identified 14 hub targets, including CaMK-II alpha, PSD-95, GluR-1, and GluN2B, as key proteins in this process. Molecular docking results indicate that BDE-209 exhibits a stronger binding affinity to GluN2B, a subunit of the N-methyl-D-aspartate (NMDA) receptors, compared to other key targets. These findings suggest that BDE-209 may disrupt the function of GluN2B-containing NMDA receptors, potentially leading to their inhibition. Such inhibition could result in reduced excitatory neurotransmission, impairing synaptic potentiation and plasticity, and ultimately contributing to neurotoxicity.
Collapse
Affiliation(s)
- Fuat Karakuş
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Van Yuzuncu Yil University, Van, Türkiye.
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Van Yuzuncu Yil University, Tuşba-Van, 65080, Türkiye.
| | - Burak Kuzu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Van Yuzuncu Yil University, Van, Türkiye
| |
Collapse
|
26
|
Li C, Wu Y, Bao S, Li H, Xu Z, Yan J, Yu X, He L, Zhang T, Liu W, Hou S, Zhang Y, Xu J, Yan T, Wang T, Yan Y, Liu J. Photo-Switchable Supramolecular Interactions Regulate K + Transmembrane Transport and Cancer Cell Apoptosis. J Am Chem Soc 2025. [PMID: 40111177 DOI: 10.1021/jacs.4c14583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Natural channel proteins (NCPs) have numerous ion transport modes, but it remains a big challenge to replicate this trait by artificial ion transport systems. Herein, we present an azobenzene-incorporated single-chain random heteropolymers (RHPs)-derived biomimetic K+ channel P3, which can switch between three ion transport states ("ON," "Partially OFF," and "Totally OFF") in both liposomes and cancer cells. The conformational adjustments of P3 activated by light-modulating two groups of supramolecular interactions ((1) hydrogen bonding and π-π interactions; (2) host-guest interactions) realize these switches, resembling the protein mechanisms that govern activity. Underlying molecular mechanisms are the photoisomerization of azobenzene moieties in P3 and their complexation with β-cyclodextrin (β-CD), enabling the exploit of a "one stone (azobenzene moiety), two birds (supramolecular interactions)" strategy. Mechanistic investigations demonstrate that P3-induced substantial K+ efflux (a 50% drop within just 4 min) causes endoplasmic reticulum (ER) stress, intriguing Ca2+ sparks, enhanced reactive oxygen species (ROS), and finally severe mitochondria-dependent apoptosis. This NCP-like channel (P3) is expected to provide new opportunities for a deeper understanding of the internal mechanisms of NCPs, as well as for treating cancer and other diseases.
Collapse
Affiliation(s)
- Cong Li
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Yaqi Wu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Sheng Bao
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Hui Li
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhengwei Xu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Jing Yan
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Xiaoxuan Yu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Lei He
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Tianlong Zhang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Wang Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Shida Hou
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Yang Zhang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Jiayun Xu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Tengfei Yan
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Tingting Wang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Yi Yan
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| | - Junqiu Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an 710129, China
| |
Collapse
|
27
|
Miguez-Cabello F, Wang XT, Yan Y, Brake N, Alexander RPD, Perozzo AM, Khadra A, Bowie D. GluA2-containing AMPA receptors form a continuum of Ca 2+-permeable channels. Nature 2025:10.1038/s41586-025-08736-2. [PMID: 40108453 DOI: 10.1038/s41586-025-08736-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025]
Abstract
Fast excitatory neurotransmission in the mammalian brain is mediated by cation-selective AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors (AMPARs)1. AMPARs are critical for the learning and memory mechanisms of Hebbian plasticity2 and glutamatergic synapse homeostasis3, with recent work establishing that AMPAR missense mutations can cause autism and intellectual disability4-7. AMPARs have been grouped into two functionally distinct tetrameric assemblies based on the inclusion or exclusion of the GluA2 subunit that determines Ca2+ permeability through RNA editing8,9. GluA2-containing AMPARs are the most abundant in the central nervous system and considered to be Ca2+ impermeable10. Here we show this is not the case. Contrary to conventional understanding, GluA2-containing AMPARs form a continuum of polyamine-insensitive ion channels with varying degrees of Ca2+ permeability. Their ability to transport Ca2+ is shaped by the subunit composition of AMPAR tetramers as well as the spatial orientation of transmembrane AMPAR regulatory proteins and cornichon auxiliary subunits. Ca2+ crosses the ion-conduction pathway by docking to an extracellular binding site that helps funnel divalent ions into the pore selectivity filter. The dynamic range in Ca2+ permeability, however, arises because auxiliary subunits primarily modify the selectivity filter. Taken together, our work proposes a broader role for AMPARs in Ca2+ signalling in the mammalian brain and offers mechanistic insight into the pathogenic nature of missense mutations.
Collapse
Affiliation(s)
| | - Xin-Tong Wang
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Yuhao Yan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Niklas Brake
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- Quantitative Life Sciences PhD program, McGill University, Montreal, Quebec, Canada
| | - Ryan P D Alexander
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Amanda M Perozzo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Anmar Khadra
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Derek Bowie
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
28
|
Bosco J, Gagliano E, Boshae KL, Statz JP, Wheeler TB, Cuello D, Sliter A, Newby C, Lin B, Demeler A, Pierpont CL, Yates-Hansen C, Sydor MJ, Ferrini ME, Kuch KC, Cooper BS, Piggott BJ, Certel SJ, Hansen KB, Sprang SR, Bowler B, McClelland L, Berkmen M, Voronina E. A galactose-based auto-expression system improves T7-inducible protein production in Escherichia coli. Sci Rep 2025; 15:8936. [PMID: 40089537 PMCID: PMC11910606 DOI: 10.1038/s41598-025-91954-5] [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: 10/10/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
Protein production using Escherichia coli is a cornerstone of modern biotechnology. In this study, we developed a novel auto-expression medium to maximize protein production. Each E. coli strain tested was capable of auto-expression in response to galactose, including strains in which the endogenous lacZ had been disrupted. This provides key evidence that galactose can regulate the lac operon independent of known lac operon-regulated metabolism. The enhanced capabilities of the novel auto-expression medium were documented across protein production systems including (1) increased yields for routinely expressed proteins (e.g. eGFP), (2) improved expression of human cytochrome c within a dual expression system, (3) robust auto-expression in lacZ-deficient strains producing proteins with challenging disulfide bonds, and (4) reproducible 8-fold increase in SpCas9 yields, at ≥ 95% purity. This novel medium can streamline production and improve yields for routine as well as challenging proteins, accelerating recombinant protein production and creating new opportunities in biotechnology and structural biology.
Collapse
Affiliation(s)
- James Bosco
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA.
| | - Emily Gagliano
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kassandra L Boshae
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - John P Statz
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Timothy B Wheeler
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - DeAnna Cuello
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Ashlyn Sliter
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Christian Newby
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Bernice Lin
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Aysha Demeler
- Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
| | - C Logan Pierpont
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Cindee Yates-Hansen
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Matthew J Sydor
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Maria E Ferrini
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kellie C Kuch
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Brandon S Cooper
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Beverly J Piggott
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Sarah J Certel
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kasper B Hansen
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Stephen R Sprang
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Bruce Bowler
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Levi McClelland
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Mehmet Berkmen
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Ekaterina Voronina
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA.
| |
Collapse
|
29
|
Banzato M, Furlan A, Locatelli P, Sgrignani J, Ongaro A, Dolmella A, De Martin S, Comai S, Cavalli A, Inturrisi C, Bettini E, Manfredi PL, Mattarei A. New Synthesis and Pharmacological Evaluation of Enantiomerically Pure ( R)- and ( S)-Methadone Metabolites as N-Methyl-d-aspartate Receptor Antagonists. J Med Chem 2025; 68:5455-5470. [PMID: 39999356 PMCID: PMC11912475 DOI: 10.1021/acs.jmedchem.4c02605] [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: 02/27/2025]
Abstract
N-Methyl-d-aspartate receptor (NMDAR) is gaining increasing interest as a pharmacological target for the development of fast-acting antidepressants. (S)-Methadone (esmethadone), has recently shown promising efficacy for the treatment of major depressive disorder. However, methods for its enantiopure preparation still rely on complex and expensive resolution procedures. In addition, enantiopure methadone metabolites have never been evaluated for their NMDAR activity. Here, we report the development of a novel chiral pool approach, based on cyclic sulfamidate ring-opening reaction, for the asymmetric synthesis of (R)- and (S)-methadone, and the application of this methodology to the stereodivergent synthesis of 20 enantiopure methadone metabolites. The compounds were evaluated for their NMDAR antagonism and for their affinity toward a series of relevant CNS receptors. Strikingly, N-demethylated (6R)-methadol metabolites retain the higher NMDAR uncompetitive antagonism of (R)-methadone, while presenting lower opioid receptor affinity compared to (S)-methadone. These compounds could represent novel candidates for drug development in CNS disorders.
Collapse
Affiliation(s)
- Marco Banzato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Francesco Marzolo 5, 35131 Padua, Italy
| | - Alberto Furlan
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Francesco Marzolo 5, 35131 Padua, Italy
- Institute for Research in Biomedicine, Via Chiesa 5, 6500 Bellinzona, Switzerland
| | - Patrizia Locatelli
- Institute for Research in Biomedicine, Via Chiesa 5, 6500 Bellinzona, Switzerland
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine, Via Chiesa 5, 6500 Bellinzona, Switzerland
| | - Alberto Ongaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Francesco Marzolo 5, 35131 Padua, Italy
| | - Alessandro Dolmella
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Francesco Marzolo 5, 35131 Padua, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Francesco Marzolo 5, 35131 Padua, Italy
| | - Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Francesco Marzolo 5, 35131 Padua, Italy
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padua, Italy
- Department of Psychiatry, McGill University, 1033 Pine Avenue West, Montreal, Quebec H3A 1A1, Canada
- IRCSS San Raffaele Scientific Institute, via Olgettina 58, 20132 Milan, Italy
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Via Chiesa 5, 6500 Bellinzona, Switzerland
| | | | - Ezio Bettini
- In Vitro Pharmacology Department, Aptuit, An Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Paolo L Manfredi
- Relmada Therapeutics, Coral Gables, Florida 33134, United States
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Francesco Marzolo 5, 35131 Padua, Italy
| |
Collapse
|
30
|
Li N, Wei Y, Li R, Meng Y, Zhao J, Bai Q, Wang G, Zhao Y. Modulation of the human GlyT1 by clinical drugs and cholesterol. Nat Commun 2025; 16:2412. [PMID: 40069141 PMCID: PMC11897355 DOI: 10.1038/s41467-025-57613-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/21/2025] [Indexed: 03/15/2025] Open
Abstract
Glycine transporter 1 (GlyT1) is a key player in shaping extracellular glutamatergic signaling processes and holds promise for treating cognitive impairments associated with schizophrenia by inhibiting its activity and thus enhancing the function of NMDA receptors. Despite its significant role in physiological and pharmacology, its modulation mechanism by clinical drugs and internal lipids remains elusive. Here, we determine cryo-EM structures of GlyT1 in its apo state and in complex with clinical trial drugs iclepertin and sarcosine. The GlyT1 in its apo state is determined in three distinct conformations, exhibiting a conformational equilibrium of the transport cycle. The complex structures with inhibitor iclepertin and sarcosine elucidate their unique binding poses with GlyT1. Three binding sites of cholesterol are determined in GlyT1, two of which are conformation-dependent. Transport kinetics studies reveal that a delicate binding equilibrium for cholesterol is crucial for the conformational transition of GlyT1. This study significantly enhances our understanding of the physiological and pharmacological aspects of GlyT1.
Collapse
Affiliation(s)
- Na Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yiqing Wei
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Renjie Li
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yufei Meng
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jun Zhao
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang, Shandong, China
| | - Qinru Bai
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Gang Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| | - Yan Zhao
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
31
|
Denley MCS, Straub MS, Marcionelli G, Güra MA, Penton D, Delvendahl I, Poms M, Vekeriotaite B, Cherkaoui S, Conte F, von Meyenn F, Froese DS, Baumgartner MR. Mitochondrial dysfunction drives a neuronal exhaustion phenotype in methylmalonic aciduria. Commun Biol 2025; 8:410. [PMID: 40069408 PMCID: PMC11897345 DOI: 10.1038/s42003-025-07828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
Methylmalonic aciduria (MMA) is an inborn error of metabolism resulting in loss of function of the enzyme methylmalonyl-CoA mutase (MMUT). Despite acute and persistent neurological symptoms, the pathogenesis of MMA in the central nervous system is poorly understood, which has contributed to a dearth of effective brain specific treatments. Here we utilised patient-derived induced pluripotent stem cells and in vitro differentiation to generate a human neuronal model of MMA. We reveal strong evidence of mitochondrial dysfunction caused by deficiency of MMUT in patient neurons. By employing patch-clamp electrophysiology, targeted metabolomics, and bulk transcriptomics, we expose an altered state of excitability, which is exacerbated by application of dimethyl-2-oxoglutarate, and we suggest may be connected to metabolic rewiring. Our work provides first evidence of mitochondrial driven neuronal dysfunction in MMA, which through our comprehensive characterisation of this paradigmatic model, enables first steps to identifying effective therapies.
Collapse
Affiliation(s)
- Matthew C S Denley
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Monique S Straub
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Giulio Marcionelli
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Miriam A Güra
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - David Penton
- Electrophysiology Core Facility, University of Zurich, Zurich, CH-8057, Switzerland
| | - Igor Delvendahl
- Department of Molecular Life Sciences, University of Zurich, Zurich, CH-8057, Switzerland
| | - Martin Poms
- Clinical Chemistry and Biochemistry and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Beata Vekeriotaite
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, CH-8603, Switzerland
| | - Sarah Cherkaoui
- Pediatric Cancer Metabolism Laboratory, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Federica Conte
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud, University Medical Center, Nijmegen, 6525 GA, Netherlands
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, CH-8603, Switzerland
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland.
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland.
| |
Collapse
|
32
|
Antal M. Molecular Anatomy of Synaptic and Extrasynaptic Neurotransmission Between Nociceptive Primary Afferents and Spinal Dorsal Horn Neurons. Int J Mol Sci 2025; 26:2356. [PMID: 40076973 PMCID: PMC11900602 DOI: 10.3390/ijms26052356] [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: 12/04/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Sensory signals generated by peripheral nociceptors are transmitted by peptidergic and nonpeptidergic nociceptive primary afferents to the superficial spinal dorsal horn, where their central axon terminals establish synaptic contacts with secondary sensory spinal neurons. In the case of suprathreshold activation, the axon terminals release glutamate into the synaptic cleft and stimulate postsynaptic spinal neurons by activating glutamate receptors located on the postsynaptic membrane. When overexcitation is evoked by peripheral inflammation, neuropathy or pruritogens, peptidergic nociceptive axon terminals may corelease various neuropeptides, neurotrophins and endomorphin, together with glutamate. However, in contrast to glutamate, neuropeptides, neurotrophins and endomorphin are released extrasynaptically. They diffuse from the site of release and modulate the function of spinal neurons via volume transmission, activating specific extrasynaptic receptors. Thus, the released neuropeptides, neurotrophins and endomorphin may evoke excitation, disinhibition or inhibition in various spinal neuronal populations, and together with glutamate, induce overall overexcitation, called central sensitization. In addition, the synaptic and extrasynaptic release of neurotransmitters is subjected to strong retrograde control mediated by various retrogradely acting transmitters, messengers, and their presynaptic receptors. Moreover, the composition of this complex chemical apparatus is heavily dependent on the actual patterns of nociceptive primary afferent activation in the periphery. This review provides an overview of the complexity of this signaling apparatus, how nociceptive primary afferents can activate secondary sensory spinal neurons via synaptic and volume transmission in the superficial spinal dorsal horn, and how these events can be controlled by presynaptic mechanisms.
Collapse
Affiliation(s)
- Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
33
|
Zhigulin AS, Barygin OI. Mechanisms of NMDA receptor inhibition by vortioxetine - Comparison with fluoxetine. Eur J Pharmacol 2025; 998:177460. [PMID: 40049576 DOI: 10.1016/j.ejphar.2025.177460] [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: 11/05/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are involved in the pathophysiology of depression and are inhibited by many antidepressants. In this work, we studied the action of the vortioxetine, a relatively new multitarget antidepressant, on native NMDARs in rat hippocampal CA1 pyramidal neurons and compared it to the action of structurally similar antidepressant fluoxetine. Vortioxetine inhibited these receptors with IC50 value of 11 ± 1 μM at -80 mV holding voltage, being about three-fold more potent than fluoxetine in these conditions. The inhibition by both compounds was not competitive. Both vortioxetine and fluoxetine demonstrated complex voltage dependence with voltage-dependent and voltage-independent components. The voltage-dependent component corresponded to trapping channel block, while the voltage-independent component - to allosteric inhibition. Vortioxetine and fluoxetine were able to inhibit both open and closed NMDAR channels. Thus, NMDARs can be among important targets for vortioxetine or structurally related drugs. In addition, structural similarity of vortioxetine and fluoxetine allows to assume that these compounds may share other molecular targets besides serotonin transporter and NMDARs.
Collapse
Affiliation(s)
- Arseniy S Zhigulin
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint-Petersburg, Russia
| | - Oleg I Barygin
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint-Petersburg, Russia.
| |
Collapse
|
34
|
Tahiri E, Corti E, Duarte CB. Regulation of Synaptic NMDA Receptor Activity by Post-Translational Modifications. Neurochem Res 2025; 50:110. [PMID: 40029461 DOI: 10.1007/s11064-025-04346-6] [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: 12/03/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
NMDA receptors for the neurotransmitter glutamate are widely distributed in the central nervous system, playing important roles in brain development, function and plasticity. Alterations in their activity are also important mediators in neuropsychiatric and neurodegenerative disorders. The different NMDA receptor subunits (GluN1, GluN2A-D and GluN3A, B) share a similar structure and membrane topology, with an intracellular C-terminus tail responsible for the interaction with proteins important for the trafficking of the receptors, and to control their surface distribution and signalling activity. The latter sequence varies among subunits but consistently contains the majority of post-translational modification sites on NMDA receptors. These modifications, including phosphorylation, ubiquitination, and palmitoylation, regulate interactions with intracellular proteins. Differences in the amino acid sequence between NMDA receptor subunits lead to a differential regulation by post-translational modifications. Since NMDA receptors are formed by oligomerization of different subunits, and each subunit is regulated in a specific manner, this creates multiple possibilities for regulation of these receptors, with impact in synaptic function and plasticity. This review addresses the diversity of mechanisms involved in the post-translational modification of NMDA receptor subunits, and their impact on the activity and distribution of the receptors, as well as their function in nerve cells.
Collapse
Affiliation(s)
- Emanuel Tahiri
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CiBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- III- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Elisa Corti
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CiBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- III- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Carlos B Duarte
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CiBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, Coimbra, 3004-504, Portugal.
| |
Collapse
|
35
|
Opal P, Swanson GT. Bridging the gap: insights into sensorimotor deficits in NMDA receptor antibody encephalitis. J Clin Invest 2025; 135:e188251. [PMID: 40026244 DOI: 10.1172/jci188251] [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: 03/05/2025] Open
Abstract
N-methyl-d-aspartate (NMDA) receptor-mediated autoimmune encephalitis (NMDAR-AE) is the most common cause of autoimmune encephalitis, especially in children and young adults. The disorder is caused by antibodies directed against the GluN1 protein, an obligatory constituent of NMDA receptors, which are key signaling molecules in brain development, learning and memory, and executive function. The manuscript by Zhou et al. offers key insights into aberrant development of cortical pathways that may underly persistent sensorimotor deficits associated with this encephalitis in a newly generated mouse model. This study convincingly links transient exposure to a patient-derived anti-GluN1 mAb during a critical developmental period to lasting disruptions in interhemispheric connectivity through callosal projections. These findings provide insight into the impact of a prevalent autoimmune disorder on fundamental aspects of brain development and establish a model system that could be further employed to probe other aspects of NMDAR-AE pathogenesis.
Collapse
Affiliation(s)
- Puneet Opal
- Denning Ataxia Center, Davee Department of Neurology and Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois,USA
| | - Geoffrey T Swanson
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
36
|
Radin DP, Lippa A, Rana S, Fuller DD, Smith JL, Cerne R, Witkin JM. Amplification of the therapeutic potential of AMPA receptor potentiators from the nootropic era to today. Pharmacol Biochem Behav 2025; 248:173967. [PMID: 39894310 PMCID: PMC11849398 DOI: 10.1016/j.pbb.2025.173967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors (AMPA receptors or AMPARs) are involved in fast excitatory neurotransmission and as such control multiple important physiological processes. AMPARs also are involved in the dynamics of synaptic plasticity in the nervous system where they impact neuroplastic responses such as long-term facilitation and long-term potentiation that regulate biological functions ranging from breathing to cognition. AMPARs also regulate neurotrophic factors that are strategically involved in neural plastic changes in the nervous system. As with other major ionotropic receptors, modulation of AMPARs can have prominent effects on biological systems that can include marked tolerability issues. AMPAR potentiators (AMPAkines) are positive allosteric modulators of AMPARs which have therapeutic potential. Medicinal chemistry combined with new pharmacological findings have defined AMPAkines with favorable oral bioavailability and pharmacological safety parameters that enable clinical advancement of their therapeutic utility. AMPAkines are being investigated in patients with diverse neurological and psychiatric disorders including spinal cord injury (breathing and bladder function), cognition, attention-deficit-hyperactivity disorder, and major depressive disorder. The present discussion of this class of compounds focuses on their general value as therapeutics through their impact on synaptic plasticity.
Collapse
Affiliation(s)
| | - Arnold Lippa
- RespireRx Pharmaceuticals Inc., Glen Rock, NJ, USA
| | - Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery St. Vincent's Hospital, Indianapolis, IN, USA
| | - Rok Cerne
- RespireRx Pharmaceuticals Inc., Glen Rock, NJ, USA; Laboratory of Antiepileptic Drug Discovery St. Vincent's Hospital, Indianapolis, IN, USA; Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, Ljubljana, Slovenia
| | - Jeffrey M Witkin
- RespireRx Pharmaceuticals Inc., Glen Rock, NJ, USA; Laboratory of Antiepileptic Drug Discovery St. Vincent's Hospital, Indianapolis, IN, USA; Department of Neurosciene and Trauma Research, Ascension St. Vincent Hospital, Indianapolis IN, USA.
| |
Collapse
|
37
|
Blicharz-Futera K, Kamiński M, Grychowska K, Canale V, Zajdel P. Current development in sulfonamide derivatives to enable CNS-drug discovery. Bioorg Chem 2025; 156:108076. [PMID: 39889550 DOI: 10.1016/j.bioorg.2024.108076] [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: 10/06/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 02/03/2025]
Abstract
The encouraging therapeutic potential of sulfonamide-based derivatives has been unraveled by breakthrough discovery of Paul Ehrlich, who pointed out the possibility of fighting microbes with chemicals. Over the decades, the utility of sulfonamides has expanded beyond antimicrobial agents, revealing their usefulness in many areas of pharmacotherapy, including the treatment of central nervous system (CNS) diseases. Through a detailed analysis of preclinical and clinical data, we identify key sulfonamide-based compounds that have demonstrated significant CNS activity. We also discuss the challenges in the development of sulfonamide derivatives as enzyme/ion channel inhibitors or receptor ligands for CNS applications, describing their mode of action and therapeutic significance. This is followed by the characteristics of pharmacological targets, structure-activity relationships, ADMET properties, efficacy in experimental animal models, and outcomes from clinical trials. Overall, the versatile nature of arylsulfonamides makes them a valuable motif in drug discovery, offering diverse opportunities for the development of novel agents for treating CNS disorders.
Collapse
Affiliation(s)
- Klaudia Blicharz-Futera
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Łazarza Street, 31-530 Krakow, Poland
| | - Michał Kamiński
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Łazarza Street, 31-530 Krakow, Poland
| | - Katarzyna Grychowska
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Vittorio Canale
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Paweł Zajdel
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland.
| |
Collapse
|
38
|
Garcia-Bonilla L. NP10679: A new horizon for neuroprotection in aneurysmal subarachnoid hemorrhage. J Pharmacol Exp Ther 2025; 392:103390. [PMID: 39933230 DOI: 10.1016/j.jpet.2025.103390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Affiliation(s)
- Lidia Garcia-Bonilla
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
39
|
Santos‐Gómez A, Juliá‐Palacios N, Rejano‐Bosch A, Marí‐Vico R, Miguez‐Cabello F, Masana M, Soto D, Olivella M, García‐Cazorla À, Altafaj X. Spermidine Treatment Improves GRIN2B Loss-Of-Function, A Primary Disorder of Glutamatergic Neurotransmission. J Inherit Metab Dis 2025; 48:e70015. [PMID: 40024627 PMCID: PMC11872566 DOI: 10.1002/jimd.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/22/2025] [Accepted: 02/14/2025] [Indexed: 03/04/2025]
Abstract
GRIN-related disorders (GRD) developmental and epileptic encephalopathies (DEEs) display a clinical spectrum including developmental delay, hypotonia, intellectual disability, epilepsy, and autistic traits. The presence of de novo pathogenic variants in the GRIN genes alters the N-methyl D-aspartate receptor (NMDAR) function, with a genotype-phenotype relationship. Despite recent advances to elucidate GRD pathophysiological mechanisms and to find treatments, to date, GRD therapeutic arms are still scarce and with limited efficacy. Herein, we investigated whether the natural polyamine spermine-positive allosteric modulators of GluN2B subunit-containing NMDARs-or its precursor spermidine might rescue NMDAR hypofunctionality. In heterologous cell systems, administration of spermine potentiated wild-type and loss-of-function (LoF) NMDAR-mediated currents and attenuated synaptic density deficits. Functionally, the putative therapeutic benefit of spermidine (spermine precursor) was assessed in constitutive Grin2b+/- heterozygous mice, a GRIN2B-LoF genetic murine model recapitulating GRD-like synaptic, motor, and cognitive alterations. Chronic spermidine administration in young adult Grin2b+/- mice partially rescued hippocampal long-term potentiation deficits in hippocampal slices of Grin2b+/- mice, supporting the cognitive improvement observed in behavioral phenotyping. Based on these preclinical findings, a case study was conducted in two pediatric patients harboring mild GRIN2B-LoF variants. Importantly, in line with preclinical findings, 18 months of spermidine treatment resulted in the amelioration of adaptive behavior (notably in the younger treated patient), with the absence of noticeable side effects. Overall, our findings provide both preclinical and clinical data supporting the benefit of spermidine for the treatment of GRD in individuals harboring GRIN2B-LoF variants.
Collapse
Affiliation(s)
- A. Santos‐Gómez
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)University of BarcelonaBarcelonaSpain
| | - N. Juliá‐Palacios
- Neurometabolic Unit, Department of NeurologyHospital Sant Joan de Déu—CIBERERBarcelonaSpain
| | - A. Rejano‐Bosch
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)University of BarcelonaBarcelonaSpain
| | - R. Marí‐Vico
- Neurometabolic Unit, Department of NeurologyHospital Sant Joan de Déu—CIBERERBarcelonaSpain
| | - F. Miguez‐Cabello
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)University of BarcelonaBarcelonaSpain
| | - M. Masana
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)University of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - D. Soto
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)University of BarcelonaBarcelonaSpain
| | - M. Olivella
- Bioinformatics and Medical Statistics GroupUniversity of Vic—Central University of CataloniaVicSpain
- Institute for Research and Innovation in Life and Health Sciences (IRIS‐CC)University of Vic—Central University of CataloniaVicSpain
| | - À. García‐Cazorla
- Neurometabolic Unit, Department of NeurologyHospital Sant Joan de Déu—CIBERERBarcelonaSpain
| | - X. Altafaj
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)University of BarcelonaBarcelonaSpain
| |
Collapse
|
40
|
Kuang C, Shi H, Cao J, Zhou Y, Zhang H, Wang Y, Zhou J. HL-IR mediates cinnamaldehyde repellency behavior in parthenogenetic Haemaphysalis longicornis. PLoS Negl Trop Dis 2025; 19:e0012877. [PMID: 40096053 PMCID: PMC11913321 DOI: 10.1371/journal.pntd.0012877] [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: 10/10/2024] [Accepted: 01/28/2025] [Indexed: 03/19/2025] Open
Abstract
Chemical repellents against arthropods have limitations in terms of toxicity and resistance. Natural plant compounds can be utilized as alternatives for developing environmentally friendly repellents for humans and animals. A variety of plant essential oils exhibit strong repellent effects against ticks; however, the mechanisms of action against ticks remain unknown. Here, we investigated the repellency of cinnamaldehyde, a primary compound found in cinnamon oil, and demonstrated that it affected the electrophysiological responses on Haller's organs of parthenogenetic Haemaphysalis longicornis. Transcriptome data indicated that the cinnamaldehyde response was linked to ionotropic receptor (HL-IR) at various tick developmental stages. HL-IR was widely expressed in a variety of tissues and developmental stages of ticks according to RT-qPCR. In situ hybridization results showed that HL-IR was highly expressed on Haller's organs of the ticks. Microinjection of HL-IR double-stranded RNA (dsRNA) showed that reduced transcript levels led to significant decreases in the tick repellency rate from cinnamaldehyde and the EAG response of Haller's organ. Experiments using competitive fluorescence binding and mutation sites showed that 218ASN was the critical binding site for cinnamaldehyde and HL-IR. We conclude that Haller's organ of ticks expresses HL-IR, and that this interaction mediates tick-repellent behavior by binding to cinnamaldehyde.
Collapse
Affiliation(s)
- Ceyan Kuang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Han Shi
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jie Cao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yongzhi Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Houshuang Zhang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yanan Wang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
41
|
Topolski MA, Gilmore BL, Khondaker R, Michniak JA, Studtmann C, Chen Y, Wagner GN, Pozo‐Aranda AE, Farris S, Swanger SA. Input-Specific Localization of NMDA Receptor GluN2 Subunits in Thalamocortical Neurons. J Neurochem 2025; 169:e70049. [PMID: 40123534 PMCID: PMC11931474 DOI: 10.1111/jnc.70049] [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: 08/24/2024] [Revised: 02/10/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Molecular and functional diversity among synapses is generated, in part, by differential expression of neurotransmitter receptors and their associated protein complexes. N-methyl-D-aspartate receptors (NMDARs) are tetrameric ionotropic glutamate receptors that most often comprise two GluN1 and two GluN2 subunits. NMDARs generate functionally diverse synapses across neuron populations through cell-type-specific expression patterns of GluN2 subunits (GluN2A-2D), which have vastly different functional properties and distinct downstream signaling. Diverse NMDAR function has also been observed at anatomically distinct inputs to a single neuron population. However, the mechanisms that generate input-specific NMDAR function remain unknown, as few studies have investigated subcellular GluN2 subunit localization in native brain tissue. We investigated NMDAR synaptic localization in thalamocortical (TC) neurons expressing all four GluN2 subunits. Utilizing high-resolution fluorescence imaging and knockout-validated antibodies, we revealed subtype- and input-specific GluN2 localization at corticothalamic (CT) versus sensory inputs to TC neurons in 4-week-old male and female C57Bl/6J mice. GluN2B was the most abundant postsynaptic subunit across all glutamatergic synapses, followed by GluN2A and GluN2C, and GluN2D was localized to the fewest synapses. GluN2B was preferentially localized to CT synapses over sensory synapses, while GluN2A and GluN2C were more abundant at sensory inputs compared to CT inputs. Furthermore, postsynaptic scaffolding proteins PSD-95 and SAP102 were preferentially colocalized with specific GluN2 subunits, and SAP102 was more abundant at sensory synapses than PSD-95. This work indicates that TC neurons exhibit subtype- and input-specific localization of diverse NMDARs and associated scaffolding proteins that likely contribute to functional differences between CT and sensory synapses.
Collapse
Affiliation(s)
| | | | - Rabeya Khondaker
- Fralin Biomedical Research Institute at VTCRoanokeVirginiaUSA
- Graduate Program in Translational Biology, Medicine, and HealthVirginia TechBlacksburgVirginiaUSA
| | - Juliana A. Michniak
- Fralin Biomedical Research Institute at VTCRoanokeVirginiaUSA
- Department of Biological Systems EngineeringVirginia TechBlacksburgVirginiaUSA
| | - Carleigh Studtmann
- Fralin Biomedical Research Institute at VTCRoanokeVirginiaUSA
- Graduate Program in Translational Biology, Medicine, and HealthVirginia TechBlacksburgVirginiaUSA
| | - Yang Chen
- Fralin Biomedical Research Institute at VTCRoanokeVirginiaUSA
- Graduate Program in Translational Biology, Medicine, and HealthVirginia TechBlacksburgVirginiaUSA
| | - Gwen N. Wagner
- Fralin Biomedical Research Institute at VTCRoanokeVirginiaUSA
| | - Aaron E. Pozo‐Aranda
- Fralin Biomedical Research Institute at VTCRoanokeVirginiaUSA
- School of NeuroscienceVirginia TechBlacksburgVirginiaUSA
| | - Shannon Farris
- Fralin Biomedical Research Institute at VTCRoanokeVirginiaUSA
- Department of Biomedical Sciences and PathobiologyVirginia‐Maryland College of Veterinary Medicine, Virginia TechBlacksburgVirginiaUSA
- Department of Internal MedicineVirginia Tech Carilion School of MedicineRoanokeVirginiaUSA
| | - Sharon A. Swanger
- Fralin Biomedical Research Institute at VTCRoanokeVirginiaUSA
- Department of Biomedical Sciences and PathobiologyVirginia‐Maryland College of Veterinary Medicine, Virginia TechBlacksburgVirginiaUSA
- Department of Internal MedicineVirginia Tech Carilion School of MedicineRoanokeVirginiaUSA
| |
Collapse
|
42
|
Mahboob A, Fatma N, Faraz A, Pervez M, Khan MA, Husain A. Advancements in the conservation of the conformational epitope of membrane protein immunogens. Front Immunol 2025; 16:1538871. [PMID: 40093005 PMCID: PMC11906443 DOI: 10.3389/fimmu.2025.1538871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Generating antibodies targeting native membrane proteins presents various challenges because these proteins are often embedded in the lipid bilayer, possess various extracellular and intracellular domains, and undergo post-translational modifications. These properties of MPs make it challenging to preserve their stable native conformations for immunization or antibody generation outside of the membranes. In addition, MPs are often hydrophobic due to their membrane-spanning regions, making them difficult to solubilize and purify in their native form. Therefore, employing purified MPs for immunogen preparation may result in denaturation or the loss of native structure, rendering them inadequate for producing antibodies recognizing native conformations. Despite these obstacles, various new approaches have emerged to address these problems. We outline recent advancements in designing and preparing immunogens to produce antibodies targeting MPs. Strategies outlined here are relevant for producing antibodies for research, diagnostics, and therapies and designing immunogens for vaccination purposes.
Collapse
Affiliation(s)
- Aisha Mahboob
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Nishat Fatma
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Ahmed Faraz
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Muntaha Pervez
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohammad Afeef Khan
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Afzal Husain
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
43
|
Costa BM, Hines D, Phillip N, Boehringer SC, Anandakrishnan R, Council-Troche M, Davis JL. Preliminary pharmacokinetics and in vivo studies indicate analgesic and stress mitigation effects of a novel NMDA receptor modulator. J Pharmacol Exp Ther 2025; 392:103401. [PMID: 40086100 DOI: 10.1016/j.jpet.2025.103401] [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: 12/10/2024] [Revised: 01/21/2025] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
N-methyl D-aspartate receptor (NMDAR) channel blockers produce analgesic and antidepressant effects by preferentially inhibiting the GluN2D subtype at lower doses. Given the distinct physiological role of GluN2 subunits, we hypothesized that compounds capable of simultaneously modulating GluN2A and GluN2D subtypes in opposite directions could serve as effective analgesics with minimal cognitive adverse effects. In this translational study, we investigated the in vivo effects of costa NMDAR stimulator 4 (CNS4), a recently discovered glutamate concentration-dependent NMDAR modulator. Pharmacokinetic data revealed that CNS4 reaches peak plasma and brain concentrations within 0.25 hours after intraperitoneal injection, with brain concentrations reaching values up to 8.4% of those in plasma (64.9 vs 5.47 μg/mL). Preliminary results showed that CNS4, a nonopioid compound, increased escape latency in mice during a hotplate assay by 1.74-fold compared with saline. In a fear conditioning experiment, CNS4 anecdotally reduced the electric shock sensation and significantly decreased stress-related defecation (fecal pellets: males, 21 vs 1; females, 19 vs 3). CNS4 also improved hyperarousal behavior (25 vs 4 jumps), without affecting fear memory parameters such as freezing episodes, duration, or latency. CNS4 caused no changes in locomotion across 8 of 9 parameters studied. Remarkably, approximately 50 hours after fear conditioning training, CNS4 prevented stress-induced excessive sucrose drinking behavior by more than 2-fold both in male and female mice. These findings suggest that CNS4 penetrates brain tissue and produces pharmacological effects such as those of NMDAR-targeting drugs but with a distinct mechanism, avoiding the undesirable side effects typical of traditional NMDAR blockers. Therefore, CNS4 holds potential as a novel nonopioid analgesic, warranting further investigation. SIGNIFICANCE STATEMENT: N-methyl D-aspartate (NMDA)-subtype glutamate receptors are an attractive target for chronic pain and posttraumatic stress disorder treatments because they play a critical role in forming emotional memories of stressful events. In this translational pharmacology work, we demonstrate the central analgesic and stress-mitigating characteristics of a novel glutamate concentration-biased NMDA receptor modulator, costa NMDA receptor stimulator 4.
Collapse
Affiliation(s)
- Blaise M Costa
- Pharmacology Division, Edward Via Virginia College of Osteopathic Medicine, Blacksburg, Virginia; Center for One Health Research, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia; Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia.
| | - De'Yana Hines
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, Virginia
| | - Nakia Phillip
- Pharmacology Division, Edward Via Virginia College of Osteopathic Medicine, Blacksburg, Virginia
| | - Seth C Boehringer
- Pharmacology Division, Edward Via Virginia College of Osteopathic Medicine, Blacksburg, Virginia; Department of Biochemistry, Virginia Tech, Blacksburg, Virginia
| | - Ramu Anandakrishnan
- Pharmacology Division, Edward Via Virginia College of Osteopathic Medicine, Blacksburg, Virginia; Center for One Health Research, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - McAlister Council-Troche
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Jennifer L Davis
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| |
Collapse
|
44
|
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] [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
|
45
|
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
|
46
|
Choquet D, Opazo P, Zhang H. AMPA receptor diffusional trapping machinery as an early therapeutic target in neurodegenerative and neuropsychiatric disorders. Transl Neurodegener 2025; 14:8. [PMID: 39934896 DOI: 10.1186/s40035-025-00470-z] [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: 08/11/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
Over the past two decades, there has been a growing recognition of the physiological importance and pathological implications surrounding the surface diffusion of AMPA receptors (AMPARs) and their diffusional trapping at synapses. AMPAR surface diffusion entails the thermally powered random Brownian lateral movement of these receptors within the plasma membrane, facilitating dynamic exchanges between synaptic and extrasynaptic compartments. This process also enables the activity-dependent diffusional trapping and accumulation of AMPARs at synapses through transient binding to synaptic anchoring slots. Recent research highlights the critical role of synaptic recruitment of AMPARs via diffusional trapping in fundamental neural processes such as the development of the early phases of long-term potentiation (LTP), contextual fear memory, memory consolidation, and sensory input-induced cortical remapping. Furthermore, studies underscore that regulation of AMPAR diffusional trapping is altered across various neurological disease models, including Huntington's disease (HD), Alzheimer's disease (AD), and stress-related disorders like depression. Notably, pharmacological interventions aimed at correcting deficits in AMPAR diffusional trapping have demonstrated efficacy in restoring synapse numbers, LTP, and memory functions in these diverse disease models, despite their distinct pathogenic mechanisms. This review provides current insights into the molecular mechanisms underlying the dysregulation of AMPAR diffusional trapping, emphasizing its role as a converging point for multiple pathological signaling pathways. We propose that targeting AMPAR diffusional trapping represents a promising early therapeutic strategy to mitigate synaptic plasticity and memory deficits in a spectrum of brain disorders, encompassing but not limited to HD, AD, and stress-related conditions. This approach underscores an integrated therapeutic target amidst the complexity of these neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Daniel Choquet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000, Bordeaux, France
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, 33000, Bordeaux, France
| | - Patricio Opazo
- UK Dementia Research Institute, Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh, EH16 4SB, UK
| | - Hongyu Zhang
- Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.
- Mohn Research Center for the Brain, University of Bergen, 5009, Bergen, Norway.
- Department of Radiology, Haukeland University Hospital, 5021, Bergen, Norway.
| |
Collapse
|
47
|
Holter KM, Klausner M, Hite MH, Moriarty C, Barth S, Pierce B, Iannucci A, Sheffler D, Cosford N, Bimonte-Nelson H, Raab-Graham KF, Gould RW. 17β-estradiol status alters NMDAR function and antipsychotic-like activity in female rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637465. [PMID: 39990384 PMCID: PMC11844370 DOI: 10.1101/2025.02.10.637465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Low 17β-estradiol (E2) in females of reproductive age, and marked E2 decline with menopause, contributes to heightened symptom severity in schizophrenia (i.e. cognitive dysfunction) and diminished response to antipsychotic medications. However, the underlying mechanisms are unknown. N-methyl-D-aspartate receptor (NMDAR) hypofunction contributes to the pathophysiology of schizophrenia, yet impact of E2 depletion on NMDAR function is not well characterized. Quantitative electroencephalography (qEEG), specifically gamma power, is a well-established functional readout of cortical activity that is elevated in patients with schizophrenia and is sensitive to alterations in NMDAR function. Using qEEG and touchscreen cognitive assessments, present studies investigated the effects of E2 on NMDAR function by administering MK-801 (NMDAR antagonist) to ovariectomized rats with or without E2 implants (Ovx+E and Ovx, respectively). Ovx rats were more sensitive to MK-801-induced elevations in gamma power and attentional impairments compared to Ovx+E rats. Further investigation revealed these effects were mediated by reduced synaptic GluN2A expression. Consistent with clinical reports, olanzapine (second-generation antipsychotic) was less effective in mitigating MK-801-induced elevations in gamma power in Ovx rats. Lastly, we examined antipsychotic-like activity of a Group II metabotropic glutamate receptor (mGlu2/3) positive allosteric modulator (PAM), SBI-0646535, as a novel therapeutic in E2-deprived conditions. SBI-0646535 reversed MK-801-induced elevations in gamma power equally regardless of E2 status. Collectively, these studies established a relationship between E2 deprivation and NMDAR function that is in part GluN2A-dependent, supporting the notion that E2 deprivation increases susceptibility to NMDAR hypofunction. This highlights the need to examine age/hormone-specific factors when considering antipsychotic response and designing novel pharmacotherapies.
Collapse
|
48
|
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
|
49
|
Tavalin SJ. Familial Alzheimer's disease mutations in amyloid precursor protein impair calcineurin signaling to NMDA receptors. J Biol Chem 2025; 301:108147. [PMID: 39732167 PMCID: PMC11910330 DOI: 10.1016/j.jbc.2024.108147] [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: 03/24/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024] Open
Abstract
Familial Alzheimer's disease (FAD) is frequently associated with mutations in the amyloid precursor protein (APP), which are thought to lead to cognitive deficits by impairing NMDA receptor (NMDAR)-dependent forms of synaptic plasticity. Given the reliance of synaptic plasticity on NMDAR-mediated Ca2+ entry, shaping of NMDAR activity by APP and/or its disease-causing variants could provide a basis for understanding synaptic plasticity impairments associated with FAD. A region of APP (residues 639-644 within APP695) processed by the γ-secretase complex, which generates amyloid-β peptides, is a hotspot for FAD mutations. This region bears similarity to a binding motif for calcineurin (CaN), a Ca2+/calmodulin-dependent phosphatase. Interaction assays confirm that APP associates with CaN in native tissue as well as in a heterologous expression system. This capacity to bind CaN extends to APP family members amyloid precursor-like protein 1 and amyloid precursor-like protein 2 (APLP1 and APLP2, respectively). Electrophysiological analysis demonstrates that APP and its family members limit NMDAR activity, in a manner consistent with CaN-dependent regulation of NMDAR desensitization. FAD mutations, in this region of APP, impair this regulation and consequently enhance NMDAR activity. Thus, by altering the landscape for CaN regulation of NMDA receptors, FAD mutations in APP may contribute to faulty information processing by modifying the dynamic range and temporal window of a critical signal for synaptic plasticity.
Collapse
Affiliation(s)
- Steven J Tavalin
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, USA.
| |
Collapse
|
50
|
Zong P, Legere N, Feng J, Yue L. TRP Channels in Excitotoxicity. Neuroscientist 2025; 31:80-97. [PMID: 38682490 DOI: 10.1177/10738584241246530] [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] [Indexed: 05/01/2024]
Abstract
Glutamate excitotoxicity is a central mechanism contributing to cellular dysfunction and death in various neurological disorders and diseases, such as stroke, traumatic brain injury, epilepsy, schizophrenia, addiction, mood disorders, Huntington's disease, Alzheimer's disease, Parkinson's disease, multiple sclerosis, pathologic pain, and even normal aging-related changes. This detrimental effect emerges from glutamate binding to glutamate receptors, including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, N-methyl-d-aspartate receptors, kainate receptors, and GluD receptors. Thus, excitotoxicity could be prevented by targeting glutamate receptors and their downstream signaling pathways. However, almost all the glutamate receptor antagonists failed to attenuate excitotoxicity in human patients, mainly due to the limited understanding of the underlying mechanisms regulating excitotoxicity. Transient receptor potential (TRP) channels serve as ancient cellular sensors capable of detecting and responding to both external and internal stimuli. The study of human TRP channels has flourished in recent decades since the initial discovery of mammalian TRP in 1995. These channels have been found to play pivotal roles in numerous pathologic conditions, including excitotoxicity. In this review, our focus centers on exploring the intricate interactions between TRP channels and glutamate receptors in excitotoxicity.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA
| | - Nicholas Legere
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| |
Collapse
|