1
|
Xu C, Gu T, Liu B, Qu H, Liu Q, Zhang L, Yin A. Astrocytic N-myc downstream-regulated gene 2 is involved in neural injury induced by sepsis-associated encephalopathy. Exp Neurol 2025; 389:115229. [PMID: 40169107 DOI: 10.1016/j.expneurol.2025.115229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
SAE is a systemic inflammatory response syndrome resulting from severe infection, which can progress to multiorgan dysfunction and mortality. Astrocytic-specific NDRG2, a stress response gene, has been implicated in regulating astrocyte reactivity and glutamate homeostasis in various neurological disorders. In this study, we initially investigated the expression and functional role of NDRG2 in SAE. Our results demonstrated that the upregulation of NDRG2 primarily inhibited Na+/K+-ATPase β1 and EAAT2, subsequently leading to glutamate toxicity and then induced astrocyte activation, neuronal dysfunction, and cellular apoptosis, ultimately leading to cognitive impairment. The deficiency of NDRG2 significantly mitigated these detrimental changes, including astrocytic activation, impaired glutamate clearance, and cognitive deficits in SAE, partly through the modulation of Na+/K+-ATPase β1. Our findings may provide new strategies for the intervention and treatment of patients with SAE in the future.
Collapse
Affiliation(s)
- Chang Xu
- From the Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tingting Gu
- From the Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bingjie Liu
- From the Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Haoran Qu
- From the Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qingzhen Liu
- From the Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lidong Zhang
- From the Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Anqi Yin
- From the Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Pomerleau F, Sulkowski BA, Suhail C, Quintero JE, Littrell OM, Murphy MP, Huettl P, Gerhardt GA. Age-related differences in resting glutamate levels and glutamate uptake in the hippocampus and frontal cortex of C57BL/6 mice. Neurobiol Aging 2025; 150:146-156. [PMID: 40121724 PMCID: PMC11981836 DOI: 10.1016/j.neurobiolaging.2025.03.008] [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: 12/17/2024] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
In normal aging, little is known in human and animal models about functional changes to glutamate neuronal systems that may contribute to age-related cognitive differences. The present studies investigated glutamate neuronal signaling in the hippocampus (dentate gyrus) and frontal cortex (infralimbic) of young adult (3-8 months), middle-aged (10-13 months), and aged (15-27 months) male and female C57BL/6 mice using microelectrode electrode array (MEA) recording technology to measure second-by-second resting levels of glutamate in anesthetized mice. Glutamate regulation was investigated in vivo by inhibiting the uptake of glutamate by local application of the competitive non-transportable blocker of excitatory amino acid transporters DL-threo-beta-benzyloxyaspartate (TBOA). Resting levels of glutamate and TBOA-induced changes in extracellular glutamate concentration were reliably measured in the hippocampus and frontal cortex of young adult, middle-aged, and aged mice and were seen to significantly increase in aging in the hippocampus. In the frontal cortex we observed an increase only in the middle-aged animals. TBOA produced robust changes in extracellular glutamate in the hippocampus and frontal cortex which showed significant changes in the kinetics of the signals in the middle-aged mice. Interestingly, the variance of the resting glutamate levels in the hippocampus of aged female mice was greater than in aged male mice, supporting a possible age-related gender difference in glutamate function. Taken together, these data support that glutamate signaling in the hippocampus and frontal cortex of aged mice is affected in normal aging with changes in glial regulation of glutamate uptake observed from the TBOA effects in the middle-aged mice.
Collapse
Affiliation(s)
- Francois Pomerleau
- Department of Neuroscience, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Neurorestoration Center, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Center for Microelectrode Technology, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA.
| | - Brittany A Sulkowski
- Department of Pharmaceutical Sciences, University of Kentucky, 789 S. Limestone, Lexington, KY 40508, USA
| | - Cocanut Suhail
- Department of Pharmaceutical Sciences, University of Kentucky, 789 S. Limestone, Lexington, KY 40508, USA
| | - Jorge E Quintero
- Department of Neuroscience, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Department of Neurosurgery, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Neurorestoration Center, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Center for Microelectrode Technology, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA
| | - O Meagan Littrell
- Department of Neuroscience, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Neurorestoration Center, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Center for Microelectrode Technology, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA
| | - M Paul Murphy
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 S. Limestone, Lexington, KY 40508, USA; Sanders Brown Center on Aging, University of Kentucky Medical Center, 800 S. Limestone, Lexington, KY 40536, USA
| | - Peter Huettl
- Department of Neuroscience, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Neurorestoration Center, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Center for Microelectrode Technology, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA
| | - Greg A Gerhardt
- Department of Neuroscience, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Department of Neurology, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Department of Neurosurgery, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 S. Limestone, Lexington, KY 40508, USA; Neurorestoration Center, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA; Center for Microelectrode Technology, University of Kentucky Medical Center, 780 Rose St, Lexington, KY 40536-0298, USA
| |
Collapse
|
3
|
Cerqueni G, Terenzi V, Preziuso A, Serfilippi T, Piccirillo S, Di Vincenzo M, Ambrogini P, Amoroso S, Orciani M, Lariccia V, Magi S. Identification of glutamate-related disease-dependent alterations in subventricular NSCs of the 3xTg Alzheimer's disease model, could they be involved in attempting damage repair? Cell Tissue Res 2025; 400:241-253. [PMID: 39960548 DOI: 10.1007/s00441-025-03954-6] [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: 11/12/2024] [Accepted: 01/30/2025] [Indexed: 06/01/2025]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterised by several factors, such as impaired glutamate neurotransmission affecting crucial functions. Neural stem cells (NSCs) are present in the adult brains of all mammalian species and contribute to the continuous generation of neural cells throughout life. The disruption of glutamate levels during the development of AD could impact NSCs' functionality, influencing their response to the microenvironment. In this work, we isolated adult neural stem cells from both triple transgenic (3xTg)-AD mice and age-matched wild type (WT) mice in order to gather information on any differences between them, particularly concerning the potential mechanisms involved in the internalisation of glutamate and its utilisation for energy production. The 3xTg model offers the ability to recapitulate human pathology with both plaque and tangle hallmarks that are involved in the process of glutamate release. In vitro culture 3xTg NSCs showed a slight morphological difference compared to WT cells and a massive reduction of proliferation and viability. Furthermore, 3xTg NSCs displayed an increase in the expression of glutamate transporters and glutamine synthetase, while glutamate dehydrogenase did not show any reduction, which is typical in AD brains. Data obtained from this basic research study suggest a possible involvement of glutamate in the cellular energy balance, indicating an attempted response of NSCs to the cytotoxic microenvironment in the early stage of AD pathology. This finding is of great interest, as it corroborates the hypothesis that targeting the glutamatergic system could be an extremely promising strategy for new therapeutics in AD.
Collapse
Affiliation(s)
- Giorgia Cerqueni
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy.
- Department of Life Science, Health, and Health Professions, Link Campus University, Via del Casale Di San Pio V, 00165, Rome, RM, Italy.
| | - Valentina Terenzi
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Mariangela Di Vincenzo
- Department of Clinical and Molecular Sciences, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Patrizia Ambrogini
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Via Ca' Le Suore 2-4, 61029, Urbino, PU, Italy
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy.
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, 60126, Ancona, AN, Italy
| |
Collapse
|
4
|
Srivastava I, Goikolea J, Ayberk Kaya T, Latorre-Leal M, Eroli F, Pereira Iglesias M, Álvarez-Jiménez L, Arroyo-García LE, Shimozawa M, Nilsson P, Fisahn A, Lindskog M, Maioli S, Loera-Valencia R. Reactive Astrocytes with Reduced Function of Glutamate Transporters in the AppNL-G-F Knock-in Mice. ACS Chem Neurosci 2025. [PMID: 40421586 DOI: 10.1021/acschemneuro.4c00714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Alzheimer's disease (AD) is associated with synaptic and memory dysfunction. One of the hallmarks of AD is reactive astrogliosis, with reactive astrocytes surrounding amyloid plaques in the brain. Astrocytes have also been shown to be actively involved in disease progression, nevertheless, mechanistic information about their role in synaptic transmission during AD pathology is lacking. Astrocytes maintain synaptic transmission by taking up extracellular glutamate during synaptic activity through astrocytic glutamate transporter GLT-1, but its function has been difficult to measure in real-time in AD pathology. Here, we used an App knock-in AD model (AppNL-G-F) carrying the Swedish, Arctic and Beyreuther mutations associated with AD and exhibiting AD-like Aβ plaque deposition and memory impairment. Using immunohistochemistry, patch-clamp of astrocytes, and Western blot from tissue and FACS isolated synaptosomes, we found that AppNL-G-F mice at 6-8 months of age have astrocytes with clearly altered morphology compared to wild-type (WT). Moreover, astrocyte glutamate clearance function in AppNL-G-F mice, measured as electrophysiological recordings of glutamate transporter currents, was severely impaired compared to WT animals. The reduction of glutamate uptake by astrocytes cannot be explained by GLT-1 protein levels, which were unchanged in synaptosomes and hippocampus of AppNL-G-F mice. Our data suggest that astrocytic glutamate transporters are affected by excess Aβ42 in the brain contributing to synaptic dysfunction in the hippocampus. This data contributes to the notion of restoring astrocyte synaptic function as a potential therapeutic strategy to treat AD.
Collapse
Affiliation(s)
- Ipsit Srivastava
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Julen Goikolea
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Tamer Ayberk Kaya
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - María Latorre-Leal
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Francesca Eroli
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Marta Pereira Iglesias
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Laura Álvarez-Jiménez
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Luis Enrique Arroyo-García
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Makoto Shimozawa
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Per Nilsson
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - André Fisahn
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Maria Lindskog
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Silvia Maioli
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
| | - Raúl Loera-Valencia
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Solna, Sweden
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Campus Chihuahua, Av. H. Colegio Militar 4700, Nombre de Dios, 31150 Chihuahua, Chih. Mexico
| |
Collapse
|
5
|
Kim SG. Nonessential amino acid is not nonessential in geriatric patients: implications for maxillofacial wound healing and bone repair. Maxillofac Plast Reconstr Surg 2025; 47:12. [PMID: 40418369 DOI: 10.1186/s40902-025-00465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Nonessential amino acids (NEAAs) are traditionally regarded as dispensable because they can be synthesized endogenously from glucose-derived intermediates. Emerging evidence, however, shows that the capacity for de novo NEAA biosynthesis declines in aged tissues, rendering several of these molecules conditionally essential during periods of stress such as surgery or fracture repair. MAIN BODY In the cranio-maxillofacial arena - where bone and soft-tissue regeneration must occur in an environment already compromised by osteoporosis, multimorbidity, and restricted oral intake - insufficient NEAA supply may translate into delayed union, wound dehiscence, and heightened infection risk. This narrative review integrates biochemical, preclinical, and clinical data to map age-dependent changes in the serine/glycine, glutamine/glutamate, arginine/citrulline, cysteine/trans-sulfuration, and alanine cycles, examines their impact on osteogenesis and mucosal healing, and evaluates nutritional or pharmacological strategies to restore NEAA sufficiency. Particular attention is paid to serine-one-carbon metabolism, the intestinal-renal arginine axis, and redox-sensitive cysteine pathways, all of which are intimately linked to collagen deposition, osteoblast differentiation, and immune modulation. CONCLUSION We conclude that proactive optimization of NEAA status - through targeted supplementation or metabolic activation - represents a low-risk, biologically rational adjunct to enhance postoperative outcomes in geriatric maxillofacial patients.
Collapse
Affiliation(s)
- Seong-Gon Kim
- Gangneung-Wonju National University, Gangneung, Republic of Korea.
| |
Collapse
|
6
|
Nielsen BS, Larsen BR, Ghazal AB, Katz A, Brennan KC, Karlish SJD, MacAulay N. Glial Versus Neuronal Na +/K +-ATPase in Activity-Evoked K + Clearance and Their Sensitivity to Elevated Extracellular K . Glia 2025. [PMID: 40387502 DOI: 10.1002/glia.70034] [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: 03/25/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
Neuronal activity in the central nervous system is associated with a [K+]o transient that is swiftly cleared from the extracellular space, predominantly by the Na+/K+-ATPase. The temporal contribution of the glial (α2β2) and the neuronal (α3β1) isoform complexes remains unresolved due to the lack of an isoform-specific inhibitor. The role of the two main brain isoform complexes in spreading depression (SD) also remains unresolved, but an SD-mediated increase in [K+]o may suppress Na+/K+-ATPase activity and thereby promote SD propagation. As demonstrated here, inhibitor assays of purified recombinant human and heterologously expressed rat Na+/K+-ATPase isoforms demonstrated significant selectivity for inhibition of α2β2 compared to α3β1 isoform complexes by a cyclobutyl perhydro-1,4-oxazepine derivative of digoxin (DcB). This phenomenon was utilized to demonstrate the temporal role of α2β2 and α3β1 in [K+]o clearance in electrically stimulated rat hippocampal slices, as monitored with ion-sensitive microelectrodes. The observations demonstrate a role of α2β2 in regulating the [K+]o during electrical stimulus of hippocampal slices, whereas α3β1 serves to restore [K+]o to baseline post-stimulus. SD can be triggered by elevated [K+]o but elevated [K+]o did not reduce the activity of the Na+/K+-ATPase or the glutamate transporters in hippocampal brain slices or upon heterologous expression of individual isoforms in Xenopus oocytes. Our results demonstrate the temporal contribution of the glial and neuronal Na+/K+-ATPase isoform complexes to clearance of [K+]o but do not support the concept that direct effects of elevated [K+]o on Na+/K+-ATPase activity or glutamate transport underlie SD propagation.
Collapse
Affiliation(s)
| | | | - Afnan Bilal Ghazal
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Adriana Katz
- Department of Molecular Biosciences, Weizmann Institute of Science, Rehovot, Israel
| | - K C Brennan
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Steven J D Karlish
- Department of Molecular Biosciences, Weizmann Institute of Science, Rehovot, Israel
| | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Wu WL, Gong XX, Qin ZH, Wang Y. Molecular mechanisms of excitotoxicity and their relevance to the pathogenesis of neurodegenerative diseases-an update. Acta Pharmacol Sin 2025:10.1038/s41401-025-01576-w. [PMID: 40389567 DOI: 10.1038/s41401-025-01576-w] [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: 10/14/2024] [Accepted: 04/26/2025] [Indexed: 05/21/2025]
Abstract
Glutamate excitotoxicity is intricately linked to the pathogenesis of neurodegenerative diseases, exerting a profound influence on cognitive functions such as learning and memory in mammals. Glutamate, while crucial for these processes, can lead to neuronal damage and death when present in excessive amounts. Our previous review delved into the cascade of excitotoxic injury events and the underlying mechanisms of excitotoxicity. Building on that foundation, this update summarizes the latest research on the role of excitotoxicity in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, as well as new cutting-edge techniques applied in the study of excitotoxicity. We also explore the mechanisms of action of various excitotoxicity inhibitors and their clinical development status. This comprehensive analysis aims to enhance our understanding of the nexus between excitotoxicity and neurodegenerative diseases, offering valuable insights for therapeutic strategies in these conditions.
Collapse
Affiliation(s)
- Wei-Long Wu
- Department of Pharmacology, College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, Soochow University, Suzhou, 215123, China
| | - Xiao-Xi Gong
- Department of Pharmacology, College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, Soochow University, Suzhou, 215123, China
| | - Zheng-Hong Qin
- Department of Pharmacology, College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, Soochow University, Suzhou, 215123, China
| | - Yan Wang
- Department of Pharmacology, College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
8
|
Beckers P, Charlier M, Azria-Richter L, Braconnier P, Desmet N, Massie A, Hermans E. Implication of system x c- in complete Freund's adjuvant-induced peripheral inflammation and associated nociceptive sensitization. Neuropharmacology 2025; 269:110340. [PMID: 39889848 DOI: 10.1016/j.neuropharm.2025.110340] [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: 11/22/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Persistent inflammation leading to neuronal sensitization in pain pathways, are key features of chronic inflammatory pain. Alike macrophages in the periphery, glial cells exacerbate hypersensitivity by releasing proalgesic mediators in the central nervous system. Expressed by peripheral and central immune cells, the cystine-glutamate antiporter system xc- plays a significant role in inflammatory responses, but its involvement in chronic inflammatory pain remains underexplored. We herein investigated the contribution of this exchanger in nociceptive hypersensitivity triggered by a peripheral inflammatory insult. METHODS Complete Freund's adjuvant (CFA) was injected into the left hind paw of wild-type C57Bl/6 female mice, of xCT-deficient mice (specific subunit of system xc-) and of mice receiving the system xc- inhibitor sulfasalazine. Paw edema was measured over three weeks and pain-associated behaviors were evaluated. Additionally, pro-inflammatory cytokine levels were assessed in blood samples. RESULTS CFA injection led to a persistent increase in paw edema and hypersensitivity to mechanical and thermal stimuli, which were less pronounced in xCT-deficient mice. This reduced sensitivity was accompanied by lower systemic pro-inflammatory cytokine levels in xCT-deficient mice. Accordingly, pharmacological inhibition of system xc- with sulfasalazine, either before or after pain induction, efficiently reduced the algesic and inflammatory responses to CFA in wild-type mice. CONCLUSION Our findings reveal a critical role for system xc- in the pathophysiology of inflammatory pain. xCT deficiency reduces pain behaviors and peripheral inflammation, positioning system xc- as a promising therapeutic target for alleviating chronic inflammatory pain.
Collapse
Affiliation(s)
- Pauline Beckers
- Institute of Neuroscience, Group of Neuropharmacology, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 53 (B1.53.01), 1200, Brussels, Belgium
| | - Mathilde Charlier
- Institute of Neuroscience, Group of Neuropharmacology, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 53 (B1.53.01), 1200, Brussels, Belgium
| | - Lorie Azria-Richter
- Institute of Neuroscience, Group of Neuropharmacology, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 53 (B1.53.01), 1200, Brussels, Belgium
| | - Pauline Braconnier
- Institute of Neuroscience, Group of Neuropharmacology, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 53 (B1.53.01), 1200, Brussels, Belgium
| | - Nathalie Desmet
- Institute of Neuroscience, Group of Neuropharmacology, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 53 (B1.53.01), 1200, Brussels, Belgium
| | - Ann Massie
- Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Emmanuel Hermans
- Institute of Neuroscience, Group of Neuropharmacology, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 53 (B1.53.01), 1200, Brussels, Belgium.
| |
Collapse
|
9
|
Das S, Mccloskey K, Nepal B, Kortagere S. EAAT2 Activation Regulates Glutamate Excitotoxicity and Reduces Impulsivity in a Rodent Model of Parkinson's Disease. Mol Neurobiol 2025; 62:5787-5803. [PMID: 39630405 PMCID: PMC11953204 DOI: 10.1007/s12035-024-04644-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/25/2024] [Indexed: 03/29/2025]
Abstract
Parkinson's disease (PD) is a systemic disease characterized by motor and nonmotor impairments. Loss of dopaminergic neurons in the substantia nigra pars compacta region in PD disrupts dopamine-glutamate homeostasis in the corticostriatal circuit, contributing to cognitive impairment. In addition, excitatory amino acid transporter-2 (EAAT2), localized predominantly to astrocytes and responsible for > 80% of synaptic glutamate clearance, is downregulated in PD, causing glutamate spillover and excitotoxicity. This altered dopamine-glutamate homeostasis and excitotoxicity may affect reward-mediated decision-making behaviors and promote impulsive behaviors in PD. In this study, we hypothesized that GTS467, a small-molecule activator of EAAT2, could effectively reduce excitotoxicity and treat cognitive impairment without promoting impulsive behavior in PD. Rats that were unilaterally lesioned with the 6-OHDA toxin to produce Parkinsonian symptoms were referred to as lesioned rats. Lesioned rats were trained to meet baseline criteria in a 5-choice serial reaction time task, and the chronic effects of GTS467 were assessed after 3 weeks of treatment. The results showed that chronic treatment with GTS467 significantly improved correct responses and reduced premature impulsive responses and omissions compared with saline treatment. This improvement in performance correlated with a reduction in glutamate levels, an increase in EAAT2 expression, and normalization of NMDA receptor subunit expression and signaling. Furthermore, transcriptomic studies on the prefrontal cortex tissue have shown the differential expression of genes involved in neuroprotection, neuroinflammation, learning, and memory. These results validate the role of glutamate excitotoxicity in promoting impulsive behaviors and suggest that GTS467 can be developed as a therapeutic agent to reduce cognitive impairment and impulsive behaviors in PD.
Collapse
Affiliation(s)
- Sanjay Das
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kyle Mccloskey
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Binod Nepal
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
10
|
Fan Y, Tian Y, Han J. The Glutamate-gated Chloride Channel Facilitates Sleep by Enhancing the Excitability of Two Pairs of Neurons in the Ventral Nerve Cord of Drosophila. Neurosci Bull 2025:10.1007/s12264-025-01397-1. [PMID: 40304877 DOI: 10.1007/s12264-025-01397-1] [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: 11/15/2024] [Accepted: 01/12/2025] [Indexed: 05/02/2025] Open
Abstract
Sleep, an essential and evolutionarily conserved behavior, is regulated by numerous neurotransmitter systems. In mammals, glutamate serves as the wake-promoting signaling agent, whereas in Drosophila, it functions as the sleep-promoting signal. However, the precise molecular and cellular mechanisms through which glutamate promotes sleep remain elusive. Our study reveals that disruption of glutamate signaling significantly diminishes nocturnal sleep, and a neural cell-specific knockdown of the glutamate-gated chloride channel (GluClα) markedly reduces nocturnal sleep. We identified two pairs of neurons in the ventral nerve cord (VNC) that receive glutamate signaling input, and the GluClα derived from these neurons is crucial for sleep promotion. Furthermore, we demonstrated that GluClα mediates the glutamate-gated inhibitory input to these VNC neurons, thereby promoting sleep. Our findings elucidate that GluClα enhances nocturnal sleep by mediating the glutamate-gated inhibitory input to two pairs of VNC neurons, providing insights into the mechanism of sleep promotion in Drosophila.
Collapse
Affiliation(s)
- Yaqian Fan
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China
| | - Yao Tian
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China.
| | - Junhai Han
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
11
|
Hodebourg R, Scofield MD, Kalivas PW, Kuhn BN. Nonneuronal contributions to synaptic function. Neuron 2025:S0896-6273(25)00260-0. [PMID: 40311612 DOI: 10.1016/j.neuron.2025.04.004] [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: 01/20/2025] [Revised: 03/09/2025] [Accepted: 04/04/2025] [Indexed: 05/03/2025]
Abstract
Synapses are elegantly integrated signaling hubs containing the canonical synaptic elements, neuronal pre- and postsynapses, along with other components of the neuropil, including perisynaptic astroglia and extracellular matrix proteins, as well as microglia and oligodendrocytes. Signaling within these multipartite hubs is essential for synaptic function and is often disrupted in neuropsychiatric disorders. We review data that have refined our understanding of how environmental stimuli shape signaling and synaptic plasticity within synapses. We propose working models that integrate what is known about how different cell types within the perisynaptic neuropil regulate synaptic functions and dysfunctions that are elicited by addictive drugs. While these working models integrate existing findings, they are constrained by a need for new technology. Accordingly, we propose directions for improving reagents and experimental approaches to better probe how signaling between cell types within perisynaptic ecosystems creates the synaptic plasticity necessary to establish and maintain adaptive and maladaptive behaviors.
Collapse
Affiliation(s)
- Ritchy Hodebourg
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Anesthesiology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, SC 29401, USA.
| | - Brittany N Kuhn
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
12
|
Alabdali A, Ben Bacha A, Alonazi M, Al-Ayadhi LY, Alanazi ASJ, El‐Ansary A. Comparative evaluation of certain biomarkers emphasizing abnormal GABA inhibitory effect and glutamate excitotoxicity in autism spectrum disorders. Front Psychiatry 2025; 16:1562631. [PMID: 40330649 PMCID: PMC12052539 DOI: 10.3389/fpsyt.2025.1562631] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/12/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social communication deficits and repetitive behaviors. An imbalance between the excitatory neurotransmitter glutamate and the inhibitory neurotransmitter gamma-aminobutyric acid (GABA) might play a crucial role in ASD. This study explores the biochemical markers associated with GABAergic and glutamatergic signaling in individuals with autism and healthy controls, aiming to identify potential diagnostic and therapeutic targets. Methods The study included 46 male individuals with autism and 26 age- and gender-matched healthy controls. The plasma levels of excitatory amino acid transporter 2 (EAAT2), potassium chloride co-transporter 2 (KCC2), Na-K-Cl co-transporter 1 (NKCC1), vitamin D3 (VD3), GABA, gamma aminobutyric acid type a receptor subunit alpha 5 (GABRA5), and glutamate were measured using ELISA. Statistical analyses, including correlation, multiple regression, and receiver operating characteristic (ROC) curve analysis, were performed to evaluate the diagnostic utility and interrelationships of these biomarkers. Results Significant biochemical differences were found between individuals with autism and healthy controls. Individuals with autism had notably lower levels of EAAT2, KCC2, NKCC1, VD3, GABA, and GABRA5, especially in the severe group. Altered KCC2/NKCC1 and GABA/glutamate ratios highlighted the imbalance in neurotransmission. The correlation and multiple regression analyses showed significant interconnections between biomarkers. The ROC analysis indicated that EAAT2, KCC2, GABA, and the ratios of KCC2/NKCC1 and GABA/glutamate have high diagnostic potential. Conclusion These findings support the hypothesis that GABA and glutamate imbalance is central to the pathophysiology of ASD. Significant disruptions in neurotransmitter signaling and chloride homeostasis, particularly in severe cases, provide insights into the neurobiological mechanisms of ASD. Restoring the GABA-glutamate balance could be an effective therapeutic strategy for ASD, warranting further research into these biochemical pathways for targeted treatments.
Collapse
Affiliation(s)
- Altaf Alabdali
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Laila Y. Al-Ayadhi
- Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | - Afaf El‐Ansary
- Autism Center, Lotus Holistic Alternative Medical Center, Abu Dhabi, United Arab Emirates
| |
Collapse
|
13
|
Badia-Soteras A, Mak A, Blok TM, Boers-Escuder C, van den Oever MC, Min R, Smit AB, Verheijen MHG. Astrocyte-synapse structural plasticity in neurodegenerative and neuropsychiatric diseases. Biol Psychiatry 2025:S0006-3223(25)01125-4. [PMID: 40254258 DOI: 10.1016/j.biopsych.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/18/2025] [Accepted: 04/11/2025] [Indexed: 04/22/2025]
Abstract
Synaptic dysfunction is a common feature across a broad spectrum of brain diseases, spanning from psychopathologies such as post-traumatic stress disorder (PTSD) and substance use disorders (SUD) to neurodegenerative diseases like Alzheimer's and Parkinson's disease (AD and PD). While neuroscience research aiming to understand the mechanisms underlying synaptic dysfunction has traditionally focused on the neuronal elements of the synapse, recent research increasingly acknowledges the contribution of astrocytes as a third element controlling synaptic transmission. This also sparked interest to investigate the tripartite synapse and its role in the etiology of neurological diseases. According to recent evidence, changes in the structural interaction between astrocytes and synapses not only play a pivotal role in modulating synaptic function and behavioral states, but are also implicated in the initiation and progression of various brain diseases. This review aims to integrate recent findings that provide insight into the molecular mechanisms underpinning astrocytic structural changes at the synapse. We offer a comprehensive discussion of the potential implications of compromised astrocyte-synapse interactions, and put forward that astrocytic synaptic coverage is generally reduced in numerous neurological disorders, with the extent of it being disease- and stage- specific. Finally, we propose outstanding questions on astrocyte-synapse structural plasticity that are relevant for future therapeutic strategies to tackle neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Aina Badia-Soteras
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands; Department of Brain Scienes, Imperial College London, London , United Kingdom; UK Dementia Research Institute at Imperial College London, London , United Kingdom
| | - Aline Mak
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Thomas M Blok
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Cristina Boers-Escuder
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Rogier Min
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam, University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Mark H G Verheijen
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
El Asmar K, El Ghoul T, Colle R, Assaf R, Martin S, Trabado S, Feve B, Verstuyft C, Becquemont L, Corruble E. Blood glutamate levels in major depression with antidepressants and benzodiazepines: A 6-month prospective cohort study. Therapie 2025:S0040-5957(25)00061-7. [PMID: 40316495 DOI: 10.1016/j.therap.2025.04.003] [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: 12/12/2024] [Revised: 03/21/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025]
Abstract
AIM OF THE STUDY Our study aims at analyzing blood glutamate levels at three time points in patients on antidepressants and benzodiazepines across different subgroups. METHODS In the 6-month METADAP cohort study, blood glutamate levels were measured in 60 patients with major depression at baseline, 3 months (M3), and 6 months (M6) after starting antidepressant treatment. All patients received a co-prescription with benzodiazepines. Mixed-effect linear regression models were used to analyze the correlation of glutamate levels over time among responders, non-responders, remitters, and non-remitters, adjusting for multiple confounders. RESULTS Glutamate blood levels showed a significant decrease. Glutamate levels decreased from M0 to M3 across all subgroups. Responders and remitters showed a further decline from M3 to M6 while they increased non-significantly from M3 to M6 among non-responders and non-remitters. Responders showed a decrease of 3.38μmol/L [95% CI (-4.94; -1.82), P<0.001], while remitters showed a decrease of 1.34μmol/L [95% CI (-2.41; -0.27), P=0.014]. CONCLUSION Glutamate blood levels decreased from M0 to M3 across all groups but varied from M3 to M6: responders and remitters continued to decline, while non-responders and non-remitters showed a non-significant increase. These results should be replicated and further explained.
Collapse
Affiliation(s)
- Kahlil El Asmar
- Inserm UMR 1018, CESP, MOODS Team, Faculté de Médecine, University Paris-Saclay, 94275 Le Kremlin-Bicêtre, France; Department of Epidemiology and Population Health, Faculty of Health Sciences, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Tala El Ghoul
- Department of Epidemiology and Population Health, Faculty of Health Sciences, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Romain Colle
- Inserm UMR 1018, CESP, MOODS Team, Faculté de Médecine, University Paris-Saclay, 94275 Le Kremlin-Bicêtre, France; Service hospitalo-universitaire de psychiatrie de Bicêtre, hôpital de Bicêtre, hôpitaux universitaires Paris-Saclay, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Rida Assaf
- Department of Computer Science, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Séverine Martin
- Inserm UMR 1018, CESP, MOODS Team, Faculté de Médecine, University Paris-Saclay, 94275 Le Kremlin-Bicêtre, France; Service hospitalo-universitaire de psychiatrie de Bicêtre, hôpital de Bicêtre, hôpitaux universitaires Paris-Saclay, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Séverine Trabado
- Inserm UMR-S U1185, faculté de médecine, université Paris-Saclay, 94275 Le Kremlin-Bicêtre, France; Service de génétique moléculaire, pharmacogénétique et hormonologie de Bicêtre, hôpital de Bicêtre, hôpitaux universitaires Paris-Saclay, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Bruno Feve
- Inserm, service d'endocrinologie, centre de recherche Saint-Antoine, hôpital Saint-Antoine, institut hospitalo-universitaire ICAN, Sorbonne université, Assistance publique-Hôpitaux de Paris, 75012 Paris, France
| | - Céline Verstuyft
- Inserm UMR 1018, CESP, MOODS Team, Faculté de Médecine, University Paris-Saclay, 94275 Le Kremlin-Bicêtre, France; Service de génétique moléculaire, pharmacogénétique et hormonologie de Bicêtre, hôpital de Bicêtre, hôpitaux universitaires Paris-Saclay, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Laurent Becquemont
- Inserm UMR 1018, CESP, MOODS Team, Faculté de Médecine, University Paris-Saclay, 94275 Le Kremlin-Bicêtre, France; Centre de recherche clinique, hôpital de Bicêtre, hôpitaux universitaires Paris-Saclay, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Emmanuelle Corruble
- Inserm UMR 1018, CESP, MOODS Team, Faculté de Médecine, University Paris-Saclay, 94275 Le Kremlin-Bicêtre, France; Service hospitalo-universitaire de psychiatrie de Bicêtre, hôpital de Bicêtre, hôpitaux universitaires Paris-Saclay, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| |
Collapse
|
15
|
Rana A, Katiyar A, Arun A, Berrios JN, Kumar G. Natural sulfur compounds in mental health and neurological disorders: insights from observational and intervention studies. Front Nutr 2025; 12:1534000. [PMID: 40271431 PMCID: PMC12014460 DOI: 10.3389/fnut.2025.1534000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/11/2025] [Indexed: 04/25/2025] Open
Abstract
Over the years, the global disease burden of neurological disorders (NDs) and mental disorders (MDs) has significantly increased, making them one of the most critical concerns and challenges to human health. In pursuit of novel therapies against MD and ND, there has been a growing focus on nutrition and health. Dietary sulfur, primarily derived from various natural sources, plays a crucial role in numerous physiological processes, including brain function. This review offers an overview of the chemical composition of several natural sources of the sulfur-rich substances such as isothiocyanates, sulforaphane, glutathione, taurine, sulfated polysaccharides, allyl sulfides, and sulfur-containing amino acids, all of which have neuroprotective properties. A multitude of studies have documented that consuming foods that are high in sulfur enhances brain function by improving cognitive parameters and reduces the severity of neuropathology by exhibiting antioxidant and anti-inflammatory properties at the molecular level. In addition, the growing role of natural sulfur compounds in repairing endothelial dysfunction, compromising blood-brain barrier and improving cerebral blood flow, are documented here. Furthermore, this review covers the encouraging results of supplementing sulfur-rich diets in many animal models and clinical investigations, along with their molecular targets in MD, such as schizophrenia, depression, anxiety, bipolar disorder, and autism spectrum disorder, and ND, such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic Lateral Sclerosis (ALS), and Multiple Sclerosis (MS). The prospects of natural sulfur compounds show great promise as they have potential applications in nutraceuticals, medicines, and functional foods to enhance brain function and prevent diseases. However, additional research is required to clarify the mechanisms by which it works, enhance its bioavailability, and evaluate its long-term safety for broad use.
Collapse
Affiliation(s)
- Apeksha Rana
- School of Life Sciences and Biotechnology, CSJM University, Kanpur, India
| | - Ashutosh Katiyar
- School of Life Sciences and Biotechnology, CSJM University, Kanpur, India
| | - Alok Arun
- Institute of Sustainable Biotechnology, Inter American University of Puerto Rico, Barranquitas, PR, United States
- Department of Biological Sciences, California State University, Turlock, CA, United States
| | - Juan Negron Berrios
- Institute of Sustainable Biotechnology, Inter American University of Puerto Rico, Barranquitas, PR, United States
| | - Gaurav Kumar
- School of Life Sciences and Biotechnology, CSJM University, Kanpur, India
| |
Collapse
|
16
|
Dai X, Xi M, Li J. Cancer metastasis: molecular mechanisms and therapeutic interventions. MOLECULAR BIOMEDICINE 2025; 6:20. [PMID: 40192949 PMCID: PMC11977077 DOI: 10.1186/s43556-025-00261-y] [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: 09/13/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
The metastatic cascade is a complicated process where cancer cells travel across multiple organs distant from their primary site of onset. Despite the wide acceptance of the 'seed and soil' theory, mechanisms driving metastasis organotropism remain mystery. Using breast cancer of different subtypes as the disease model, we characterized the 'metastatic profile of cancer cells' and the 'redox status of the organ microenvironment' as the primary determinants of cancer metastasis organotropism. Mechanically, we identified a positive correlation between cancer metabolic plasticity and stemness, and proposed oxidative stress as the selection power of cancer cells succeeding the metastasis cascade. Therapeutically, we proposed the use of pro-oxidative therapeutics in ablating cancer cells taking advantages of this fragile moment during metastasis. We comprehensively reviewed current pro-oxidative strategies for treating cancers that cover the first line chemo- and radio-therapies, approaches relying on naturally existing power including magnetic field, electric field, light and sound, nanoparticle-based anti-cancer composites obtained through artificial design, as well as cold atmospheric plasma as an innovative pro-oxidative multi-modal modality. We discussed possible combinations of pro-oxidative approaches with existing therapeutics in oncology prior to the forecast of future research directions. This paper identified the fundamental mechanics driving metastasis organotropism and proposed intervention strategies accordingly. Insights provided here may offer clues for the design of innovative solutions that may open a new paradigm for cancer treatment.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| | - Ming Xi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Jitian Li
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Henan Province, Zhengzhou, 450000, China
| |
Collapse
|
17
|
Magro G, Laterza V, Tosto F, Torrente A. Manganese Neurotoxicity: A Comprehensive Review of Pathophysiology and Inherited and Acquired Disorders. J Xenobiot 2025; 15:54. [PMID: 40278159 PMCID: PMC12028444 DOI: 10.3390/jox15020054] [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: 03/04/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
Manganese (Mn) is an essential trace element and a cofactor for several key enzymes, such as mitochondrial superoxide dismutase. Consequently, it plays an important defense role against reactive oxygen species. Despite this, Mn chronic overexposure can result in a neurological disorder referred to as manganism, which shares some similarities with Parkinson's disease. Mn levels seem regulated by many transporters responsible for its uptake and efflux. These transporters play an established role in many inherited disorders of Mn metabolism and neurotoxicity. Some inherited Mn metabolism disorders, caused by mutations of SLC30A10 and SLC39A14, assume crucial importance since earlier treatment results in a better prognosis. Physicians should be familiar with the clinical presentation of these disorders as the underlying cause of dystonia/parkinsonism and look for other accompanying features, such as liver disease and polycythemia, which are typically associated with SLC30A10 mutations. This review aims to highlight the currently known Mn transporters, Mn-related neurotoxicity, and its consequences, and it provides an overview of inherited and acquired disorders of Mn metabolism. Currently available treatments are also discussed, focusing on the most frequently encountered presentations.
Collapse
Affiliation(s)
- Giuseppe Magro
- Department of Neuroscience, “Giovanni Paolo II” Hospital, Lamezia Terme, 88100 Catanzaro, Italy
| | - Vincenzo Laterza
- Department of Medical and Surgical Sciences, Institute of Neurology, Magna Graecia University, 88100 Catanzaro, Italy
| | - Federico Tosto
- Department of Neuroscience, “Giovanni Paolo II” Hospital, Lamezia Terme, 88100 Catanzaro, Italy
| | - Angelo Torrente
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics (BiND), University of Palermo, 90129 Palermo, Italy;
| |
Collapse
|
18
|
Kovermann P, Bayat A, Fenger CD, Leeuwen L, Borovikov A, Sharkov A, Levrat V, Lesca G, Perrin L, Levy J, Fahlke C, Møller RS, Jensen AA. The severity of SLC1A2-associated neurodevelopmental disorders correlates with transporter dysfunction. EBioMedicine 2025; 114:105648. [PMID: 40174554 PMCID: PMC11999296 DOI: 10.1016/j.ebiom.2025.105648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/01/2025] [Accepted: 03/02/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Excitatory amino acid transporter 2 (EAAT2) is the predominant glutamate transporter and a key mediator of excitatory neurotransmission in the human brain. Here we present a cohort of 18 individuals harbouring 13 different SLC1A2 variants, who all present with neurodevelopmental impairment with variable symptoms and disease severities, and we delineate the impact of these variants on EAAT2 function. METHODS The consequences of nine novel missense SLC1A2 variants for expression, transport and anion channel properties of EAAT2 expressed in mammalian cells were characterized by confocal microscopy, enzyme-linked immunosorbent and [3H]-D-aspartate uptake assays, and electrophysiological recordings. FINDINGS Ten of the 13 SLC1A2 variants mediated significant changes to EAAT2 expression and/or function. These molecular phenotypes were classified into three categories: overall loss-of-function (F249Sfs∗17, A432D, A439V, c.1421+1G>C), mild gain-of-anion-channel function (I276S, G360A), and mixed loss-of-transport/gain-of-anion-channel function (G82R, L85R, L85P, P289R). In contrast, L37P, H542R and I546T did not mediate significant changes to EAAT2 expression or function. Although specific clinical outcomes in individuals carrying variants within each category varied somewhat, the three categories overall translated into distinct clinical phenotypes in terms of phenotypic traits and severity. INTERPRETATION The observed associations between functional effects and clinical phenotypes produced by these variants offer valuable insights for future predictions of progression and severity of SLC1A2-associated neurodevelopmental disorders. Furthermore, these associations between variant-induced changes in EAAT2 function and phenotypic traits could assist in tailoring personalized treatments of these disorders. FUNDING This work was funded by the German Ministry of Education and Research and by the Lundbeck Foundation.
Collapse
Affiliation(s)
- Peter Kovermann
- Forschungszentrum Jülich GmbH, Institute of Biological Information Processing 1 (IBI-1), Molekular- und Zellphysiologie, Jülich D-52428, Germany
| | - Allan Bayat
- Department of Regional Health Research, University of Southern Denmark, Odense M DK-5230, Denmark; Department of Pediatrics, Danish Epilepsy Centre Filadelfia (member of ERN EpiCARE), Dianalund DK-4293, Denmark; Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre Filadelfia (member of ERN EpiCARE), Dianalund DK-4293, Denmark
| | - Christina D Fenger
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre Filadelfia (member of ERN EpiCARE), Dianalund DK-4293, Denmark; Amplexa Genetics, Odense C DK-5000, Denmark
| | - Lisette Leeuwen
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Artem Sharkov
- Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University, Moscow, Russia; Genomed Ltd., Moscow, Russia
| | - Virginie Levrat
- Service de Pédiatrie, Centre Hospitalier Annecy Genevois, Pringy, France
| | - Gaetan Lesca
- Department of Medical Genetics, University Hospital of Lyon and Claude Bernard Lyon University, Lyon, France; Pathophysiology and Genetics of Neuron and Muscle (PNMG), UCBL, CNRS UMR5261 - INSERM U1315, Lyon, France
| | - Laurence Perrin
- Department of Genetics, APHP Nord, Robert Debré University Hospital, Paris 75019, France
| | - Jonathan Levy
- Department of Genetics, APHP Nord, Robert Debré University Hospital, Paris 75019, France; Multi-site Medical Biology Laboratory SeqOIA-FMG2025, Paris 75014, France
| | - Christoph Fahlke
- Forschungszentrum Jülich GmbH, Institute of Biological Information Processing 1 (IBI-1), Molekular- und Zellphysiologie, Jülich D-52428, Germany
| | - Rikke S Møller
- Department of Regional Health Research, University of Southern Denmark, Odense M DK-5230, Denmark; Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre Filadelfia (member of ERN EpiCARE), Dianalund DK-4293, Denmark
| | - Anders A Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø DK-2100, Denmark.
| |
Collapse
|
19
|
Balog E, Jenei G, Berkó AM, Lőrinczi B, Szatmári I, Vécsei L, Toldi J, Kis Z. Age-dependent changes in NMDA-induced excitotoxicity and neuromodulatory effects of kynurenic acid and its analogue in mouse brain slices. Neurosci Lett 2025; 854:138220. [PMID: 40154656 DOI: 10.1016/j.neulet.2025.138220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 02/01/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Kynurenic acid (KYNA) is one of the main neuroprotective substances of the kynurenine pathway. KYNA plays an important role in various neurodegenerative and psychiatric diseases. Although KYNA has been shown to have neuroprotective effects, it cannot be used as a peripherally administered drug due to its poor ability to cross the blood-brain barrier. To address this limitation, chemically modified KYNA analogues are being developed: SZR72 is one such analogue and has been shown to be protective in various animal models. Glutamate-induced excitotoxicity is a key factor in many neurodegenerative diseases. Therefore, we used the N-methyl-D-aspartate (NMDA)-induced excitotoxicity model to investigate the neuromodulatory agents. Using acute hippocampal slices from mouse brains, we investigated the potential neuroprotective effect of KYNA and its analogue, SZR72 on NMDA-induced excitotoxicity across different age groups of mice. The degree of tissue damage was assessed using biochemical and histological methods. In young animals (1- and 4-week-old), NMDA treatment caused no significant changes, and the cells were found to be resistant. However, in older animals (8-week-old and 1-year-old), NMDA caused significant damage in cells and tissue structure, which was reduced by KYNA and SZR72 treatment. To our knowledge, this is the first study to compare the neuroprotective effects of KYNA and SZR72 in animals of different ages using an in vitro NMDA excitotoxicity model.
Collapse
Affiliation(s)
- Emma Balog
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary.
| | - Gyula Jenei
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Anikó Magyariné Berkó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Bálint Lőrinczi
- Institute of Pharmaceutical Chemistry, University of Szeged, Szeged, Hungary
| | - István Szatmári
- Institute of Pharmaceutical Chemistry, University of Szeged, Szeged, Hungary; HUN-REN-SZTE Stereochemistry Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary; HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Szeged, Hungary
| | - József Toldi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Zsolt Kis
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
20
|
Tao-Cheng JH. Ultrastructural characterization of peri-synaptic astrocytic processes around cerebellar Purkinje spines under resting and stimulated conditions. Mol Brain 2025; 18:28. [PMID: 40165219 PMCID: PMC11956224 DOI: 10.1186/s13041-025-01198-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025] Open
Abstract
In mammalian brains, astroglia presence near glutamatergic excitatory synapses has generated the term "tripartite" junctions, based on the close association of astrocytic processes near the active zone formed by presynaptic axonal terminal and postsynaptic dendritic spines. One major function of these astrocytic processes is to take up glutamate that spill out of the synaptic cleft during activity, via glutamate transporters located on astroglial plasma membrane. Comapred to other regions of the brain, the cerebellar Purkinje spines in the molecular layer are virtually completely ensheathed by Bergman glia, a special type of astrocyte, unique to cerebellum. The present electron microscopy study classifies these peri-synaptic astrocytic processes (PAP) ensheathing the Purkinje spine synapses into three types based on structural criteria: (1) Type 1- astrocytic process is situated at the edge of the synaptic cleft immediately next to the synaptic active zone. Under fast perfusion fixation conditions where synapses were under resting states, ~ 58% of the PAP's were scored as Type 1. The occurrence frequency of Type 1 PAP significantly decreased to 25% upon a 5-8 min delay in perfusion fixation, where synapses were under stimulated states. (2) Type 2- astrocytic process covers part of the postsynaptic membrane containing the postsynaptic density (PSD), so that this part of the PSD is separated from its presynaptic terminal. Occurrence frequency of Type 2 PAP's significantly increased from ~ 14% under fast perfusion fixation to 31% upon delayed perfusion fixation, and the average length of the PSD edge covered by astroglia increased from 41 nm to 57 nm upon delayed perfusion fixation. (3) Type 3- astrocytic process is situated some distance away from the active zone, while the presynaptic axon terminal extends to enwrap the spine beyond the active zone. Occurrence frequency of Type 3 PAP's increased from 28 to 43% upon delayed perfusion fixation, and the average length between apposed axon terminal and spine beyond the synaptic cleft significantly increased from 98 to 209 nm upon delayed perfusion fixation. Thus, upon stimulation, the tripartite synaptic junctions undergo dynamic structural changes with the astrocytic processes moving into the open cleft to cover the exposed postsynaptic membrane containing PSD, the presynaptic axon terminals extending to wrap the postsynaptic spine beyond the synaptic cleft. Both structural changes may facilitate glutamate uptake to clear the transmitter spilled out from the synaptic cleft during intense activity and prevent damage from overstimulation.
Collapse
Affiliation(s)
- Jung-Hwa Tao-Cheng
- NINDS Electron Microscopy Facility National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
21
|
Petroccione MA, Melone M, Rathwell TJ, Dwivedi N, Grienberger C, Conti F, Scimemi A. An unsuspected physiological role for mGluRIII glutamate receptors in hippocampal area CA1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646479. [PMID: 40236245 PMCID: PMC11996470 DOI: 10.1101/2025.03.31.646479] [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
Group III metabotropic glutamate receptors (mGluRIII) are expressed broadly throughout the neocortex and hippocampus but are thought to inhibit neurotransmitter release only at a subset of synapses and in a target cell- specific manner. Accordingly, previous slice physiology experiments in hippocampal area CA1 showed that mGluRIII receptors inhibit glutamate and GABA release only at excitatory and inhibitory synapses formed onto GABAergic interneurons, not onto pyramidal cells. Here, we show that the supposed target cell-specific modulation of GABA release only occurs when the extracellular calcium concentration in the recording solution is higher than its physiological concentration in the cerebrospinal fluid. Under more physiological conditions, mGluRIII receptors inhibit GABA release at synapses formed onto both interneurons and pyramidal cells but limit glutamate release only onto interneurons. This previously unrecognized form of mGluRIII-dependent, pre-synaptic modulation of inhibition onto pyramidal cells is accounted for by a reduction in the size of the readily releasable pool, mediated by protein kinase A and its vesicle-associated target proteins, synapsins. Using in vivo whole-cell recordings in behaving mice, we demonstrate that blocking mGluRIII activation in the intact CA1 network results in net effects consistent with decreased inhibition and significantly alters CA1 place cell activity. Together, these findings challenge our current understanding of the role of mGluRIII receptors in the control of synaptic transmission and encoding of spatial information in the hippocampus.
Collapse
|
22
|
O’Donovan SM, Shan D, Wu X, Choi JH, McCullumsmith RE. Dysregulated Transcript Expression but Not Function of the Glutamate Transporter EAAT2 in the Dorsolateral Prefrontal Cortex in Schizophrenia. Schizophr Bull 2025; 51:531-542. [PMID: 38825587 PMCID: PMC11908862 DOI: 10.1093/schbul/sbae092] [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] [Indexed: 06/04/2024]
Abstract
BACKGROUND Schizophrenia (SCZ) is a serious mental illness with complex pathology, including abnormalities in the glutamate system. Glutamate is rapidly removed from the synapse by excitatory amino acid transporters (EAATs). Changes in the expression and localization of the primary glutamate transporter EAAT2 are found in the brain in central nervous system (CNS) disorders including SCZ. We hypothesize that neuronal expression and function of EAAT2 are increased in the frontal cortex in subjects diagnosed with SCZ. STUDY DESIGN EAAT2 protein expression and glutamate transporter function were assayed in synaptosome preparations from the dorsolateral prefrontal cortex (DLPFC) of SCZ subjects and age- and sex-matched nonpsychiatrically ill controls. EAAT2 splice variant transcript expression was assayed in enriched populations of neurons and astrocytes from the DLPFC. Pathway analysis of publicly available transcriptomic datasets was carried out to identify biological changes associated with EAAT2 perturbation in different cell types. RESULTS We found no significant changes in EAAT2 protein expression or glutamate uptake in the DLPFC in SCZ subjects compared with controls (n = 10/group). Transcript expression of EAAT2 and signaling molecules associated with EAAT2b trafficking (CaMKIIa and DLG1) were significantly altered in enriched populations of astrocytes and pyramidal neurons (P < .05) in SCZ (n = 16/group). These changes were not associated with antipsychotic medications. Pathway analysis also identified cell-type-specific enrichment of biological pathways associated with perturbation of astrocyte (immune pathways) and neuronal (metabolic pathways) EAAT2 expression. CONCLUSIONS Overall, these data support the growing body of evidence for the role of dysregulation of the glutamate system in the pathophysiology of SCZ.
Collapse
Affiliation(s)
| | - Dan Shan
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xiaojun Wu
- Department of Neuroscience, University of Toledo, Toledo, OH, USA
| | - Jae Hyuk Choi
- Department of Neuroscience, University of Toledo, Toledo, OH, USA
| | - Robert E McCullumsmith
- Department of Neuroscience, University of Toledo, Toledo, OH, USA
- Promedica Neuroscience Institute, Toledo, OH, USA
| |
Collapse
|
23
|
Belančić A, Janković T, Gkrinia EMM, Kristić I, Rajič Bumber J, Rački V, Pilipović K, Vitezić D, Mršić-Pelčić J. Glial Cells in Spinal Muscular Atrophy: Speculations on Non-Cell-Autonomous Mechanisms and Therapeutic Implications. Neurol Int 2025; 17:41. [PMID: 40137462 PMCID: PMC11944370 DOI: 10.3390/neurolint17030041] [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: 02/09/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by homozygous deletions or mutations in the SMN1 gene, leading to progressive motor neuron degeneration. While SMA has been classically viewed as a motor neuron-autonomous disease, increasing evidence indicates a significant role of glial cells-astrocytes, microglia, oligodendrocytes, and Schwann cells-in the disease pathophysiology. Astrocytic dysfunction contributes to motor neuron vulnerability through impaired calcium homeostasis, disrupted synaptic integrity, and neurotrophic factor deficits. Microglia, through reactive gliosis and complement-mediated synaptic stripping, exacerbate neurodegeneration and neuroinflammation. Oligodendrocytes exhibit impaired differentiation and metabolic support, while Schwann cells display abnormalities in myelination, extracellular matrix composition, and neuromuscular junction maintenance, further compromising motor function. Dysregulation of pathways such as NF-κB, Notch, and JAK/STAT, alongside the upregulation of complement proteins and microRNAs, reinforces the non-cell-autonomous nature of SMA. Despite the advances in SMN-restorative therapies, they do not fully mitigate glial dysfunction. Targeting glial pathology, including modulation of reactive astrogliosis, microglial polarization, and myelination deficits, represents a critical avenue for therapeutic intervention. This review comprehensively examines the multifaceted roles of glial cells in SMA and highlights emerging glia-targeted strategies to enhance treatment efficacy and improve patient outcomes.
Collapse
Affiliation(s)
- Andrej Belančić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | | | - Iva Kristić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jelena Rajič Bumber
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Centre Rijeka, Krešimirova 42, 51000 Rijeka, Croatia;
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Dinko Vitezić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jasenka Mršić-Pelčić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| |
Collapse
|
24
|
Thangapandian S, Fakharzadeh A, Moradi M, Tajkhorshid E. Conformational free energy landscape of a glutamate transporter and microscopic details of its transport mechanism. Proc Natl Acad Sci U S A 2025; 122:e2416381122. [PMID: 40042900 PMCID: PMC11912404 DOI: 10.1073/pnas.2416381122] [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: 08/14/2024] [Accepted: 01/21/2025] [Indexed: 03/19/2025] Open
Abstract
Removing glutamate from the synaptic cleft is vital for proper function of the brain. Excitatory amino acid transporters mediate this process by uptaking the neurotransmitter from the synaptic cleft back to the cell after its release. The archaeal homolog, GltPh, an aspartate transporter from Pyrococcus horikoshii, presents the best structurally characterized model for this family of transporters. In order to transport, GltPh undergoes elevator-like conformational changes between inward-facing (IF) and outward-facing (OF) states. Here, we characterize, at an atomic level, the OF⇌IF transition of GltPh in different apo/bound states using a combination of ensemble-based enhanced sampling techniques, employing more than two thousand of coupled simulation replicas of membrane-embedded GltPh. The resulting free-energy profiles portray the transition of apo/bound states as a complex four-stage process, while sodium binding alone locks the structure in one of its states. Along the transition, the transport domain (TD) disengages from the scaffold domain (SD), allowing it to move as a piston sliding vertically with respect to the membrane during the elevator-like motion of TD. Lipid interactions with residues comprising the SD-TD interface directly influence the large-scale conformational changes and, consequently, the energetics of transport. Structural intermediates formed during the transition leak water molecules and may correlate to the uncoupled Cl- ion conductance observed experimentally in both prokaryotic and mammalian glutamate transporters. Mechanistic insights obtained from our study provide a structural framework for better development of therapeutic for neurological disorders.
Collapse
Affiliation(s)
- Sundar Thangapandian
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL61801
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Ashkan Fakharzadeh
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL61801
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Mahmoud Moradi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR72701
| | - Emad Tajkhorshid
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL61801
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL61801
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
| |
Collapse
|
25
|
Salarvandian S, Digaleh H, Khodagholi F, Javadpour P, Asadi S, Zaman AAO, Dargahi L. Harmonic activity of glutamate dehydrogenase and neuroplasticity: The impact on aging, cognitive dysfunction, and neurodegeneration. Behav Brain Res 2025; 480:115399. [PMID: 39675635 DOI: 10.1016/j.bbr.2024.115399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
In recent years, glutamate has attracted significant attention for its roles in various brain processes. However, one of its key regulators, glutamate dehydrogenase (GDH), remains understudied despite its pivotal role in several biochemical pathways. Dysfunction or dysregulation of GDH has been implicated in aging and various neurological disorders, such as Alzheimer's disease and Parkinson's disease. In this review, the impact of GDH on aging, cognitive impairment, and neurodegenerative conditions, as exemplars of the phenomena that may affected by neuroplasticity, has been reviewed. Despite extensive research on synaptic plasticity, the precise influence of GDH on brain structure and function remains undiscovered. This review of existing literature on GDH and neuroplasticity reveals diverse and occasionally conflicting effects. Future research endeavors should aim to describe the precise mechanisms by which GDH influences neuroplasticity (eg. synaptic plasticity and neurogenesis), particularly in the context of human aging and disease progression. Studies on GDH activity have been limited by factors such as insufficient sample sizes and varying experimental conditions. Researchers should focus on investigating the molecular mechanisms by which GDH modulates neuroplasticity, utilizing various animal strains and species, ages, sexes, GDH isoforms, brain regions, and cell types. Understanding GDH's role in neuroplasticity may offer innovative therapeutic strategies for neurodegenerative and psychiatric diseases, potentially slowing the aging process and promoting brain regeneration.
Collapse
Affiliation(s)
- Shakiba Salarvandian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Digaleh
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Pegah Javadpour
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sareh Asadi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ali Orang Zaman
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Onwordi EC, Whitehurst T, Shatalina E, Carr R, Mansur A, Arumuham A, Osugo M, Marques TR, Jauhar S, Gupta S, Pappa S, Mehrotra R, Ranger M, Rahaman N, Rabiner EA, Gunn RN, Natesan S, Howes OD. The relationship between cortical synaptic terminal density marker SV2A and glutamate early in the course of schizophrenia: a multimodal PET and MRS imaging study. Transl Psychiatry 2025; 15:70. [PMID: 40025026 PMCID: PMC11873237 DOI: 10.1038/s41398-025-03269-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 01/08/2025] [Accepted: 01/31/2025] [Indexed: 03/04/2025] Open
Abstract
Loss of glutamatergic terminals is hypothesised to contribute to excitation-inhibition imbalance in schizophrenia, supported by evidence that the normal positive association between glutamate concentrations and synaptic terminal density is not found in patients with chronic schizophrenia. However, it is unknown whether the relationship between synaptic terminal density and glutamate levels is altered early in the course of illness. To address this, we investigated [11C]UCB-J distribution volume ratio (DVR) and glutamatergic markers in healthy volunteers (HV) and in antipsychotic-naïve/free patients with schizophrenia (SCZ) recruited from first-episode psychosis services. Forty volunteers (HV n = 19, SCZ n = 21) underwent [11C]UCB-J positron emission tomography and proton magnetic resonance spectroscopy (1H-MRS) imaging in the anterior cingulate cortex (ACC) and left hippocampus to index [11C]UCB-J DVR and creatine-scaled glutamate (Glu/Cr) and glutamate in combination with glutamine (Glx/Cr). In the HV but not SCZ group, [11C]UCB-J DVR was significantly positively associated with Glu/Cr (Spearman's rho = 0.55, p = 0.02) and Glx/Cr (Spearman's rho = 0.73, p = 0.0004) in the ACC, and with Glu/Cr in the left hippocampus (Spearman's rho = 0.77, p = 0.0001). DVR was significantly lower in the ACC in the SCZ group compared to the HV group (Kolmogorov-Smirnov Z = 1.44, p = 0.03). Together, these findings indicate that the normal relationship between levels of a synaptic terminal density marker and levels of glutamate is disrupted early in the course of schizophrenia. This is consistent with the hypothesis that there is loss of glutamatergic terminals at illness onset.
Collapse
Affiliation(s)
- Ellis Chika Onwordi
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK.
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, UK.
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Centre for Psychiatry and Mental Health, Wolfson Institute of Population Health, Queen Mary University of London, London, UK.
- East London NHS Foundation Trust, London, UK.
| | - Thomas Whitehurst
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- City & Hackney Early and Quick Intervention in Psychosis, East London NHS Foundation Trust, London, UK
| | - Ekaterina Shatalina
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Richard Carr
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | | | - Atheeshaan Arumuham
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Martin Osugo
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Tiago Reis Marques
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Sameer Jauhar
- Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King's College, London, UK
| | - Susham Gupta
- Tower Hamlets Early Intervention Service, 51 Three Colts Lane, Bethnal Green, East London NHS Foundation Trust, London, UK
| | - Sofia Pappa
- Department of Brain Sciences, Imperial College London, The Commonwealth Building, Hammersmith Hospital, London, UK
- Research and Development Department, West London NHS Trust, London, UK
| | - Ravi Mehrotra
- Lakeside Unit, West Middlesex University Hospital, West London NHS Trust, London, UK
| | - Maja Ranger
- Westminster Community Rehabilitation Team & Bluebell Lodge, Central and North West London NHS Foundation Trust, London, UK
| | - Nikola Rahaman
- Westminster and Kensington and Chelsea Early Intervention Service, Central and North West London NHS Foundation Trust, Hathaway House, London, UK
| | - Eugenii A Rabiner
- Invicro, Burlington Danes Building, London, UK
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Roger N Gunn
- Department of Brain Sciences, Imperial College London, The Commonwealth Building, Hammersmith Hospital, London, UK
- Invicro, Burlington Danes Building, London, UK
| | - Sridhar Natesan
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Oliver D Howes
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK.
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, UK.
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| |
Collapse
|
27
|
Andersen JV. The Glutamate/GABA-Glutamine Cycle: Insights, Updates, and Advances. J Neurochem 2025; 169:e70029. [PMID: 40066661 PMCID: PMC11894596 DOI: 10.1111/jnc.70029] [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: 11/16/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Synaptic homeostasis of the principal neurotransmitters glutamate and GABA is tightly regulated by an intricate metabolic coupling between neurons and astrocytes known as the glutamate/GABA-glutamine cycle. In this cycle, astrocytes take up glutamate and GABA from the synapse and convert these neurotransmitters into glutamine. Astrocytic glutamine is subsequently transferred to neurons, serving as the principal precursor for neuronal glutamate and GABA synthesis. The glutamate/GABA-glutamine cycle integrates multiple cellular processes, including neurotransmitter release, uptake, synthesis, and metabolism. All of these processes are deeply interdependent and closely coupled to cellular energy metabolism. Astrocytes display highly active mitochondrial oxidative metabolism and several unique metabolic features, including glycogen storage and pyruvate carboxylation, which are essential to sustain continuous glutamine release. However, new roles of oligodendrocytes and microglia in neurotransmitter recycling are emerging. Malfunction of the glutamate/GABA-glutamine cycle can lead to severe synaptic disruptions and may be implicated in several brain diseases. Here, I review central aspects and recent advances of the glutamate/GABA-glutamine cycle to highlight how the cycle is functionally connected to critical brain functions and metabolism. First, an overview of glutamate, GABA, and glutamine transport is provided in relation to neurotransmitter recycling. Then, central metabolic aspects of the glutamate/GABA-glutamine cycle are reviewed, with a special emphasis on the critical metabolic roles of glial cells. Finally, I discuss how aberrant neurotransmitter recycling is linked to neurodegeneration and disease, focusing on astrocyte metabolic dysfunction and brain lipid homeostasis as emerging pathological mechanisms. Instead of viewing the glutamate/GABA-glutamine cycle as individual biochemical processes, a more holistic and integrative approach is needed to advance our understanding of how neurotransmitter recycling modulates brain function in both health and disease.
Collapse
Affiliation(s)
- Jens V. Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
28
|
Chao CK, Blecha J, Polvoy I, Nillo RM, Seo Y, Wilson DM, Forsayeth JR, VanBrocklin HF, Gerdes JM. First-in-human healthy volunteer dosimetry studies of the excitatory amino acid transporter 2 (EAAT2) PET imaging tracer methyl N 4-(7-[ 18F]fluoro-9H-fluoren-2-yl)asparaginate, [ 18F]RP-115. Nucl Med Biol 2025; 142-143:108992. [PMID: 39913962 DOI: 10.1016/j.nucmedbio.2025.108992] [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: 10/29/2024] [Revised: 12/21/2024] [Accepted: 01/08/2025] [Indexed: 05/04/2025]
Abstract
OBJECTIVE AND BACKGROUND The objective of this first-in-human study was to investigate the radiosynthesis, and the preliminary safety, biodistribution, and organ radiation dosimetry of the positron emission tomography (PET) imaging tracer methyl N4-([18F]7-fluoro-9H-fluoren-2-yl)asparaginate, known as [18F]RP-115, in a small cohort (n=8) of healthy volunteers. The [18F]RP-115 tracer is a methyl ester prodrug and undergoes metabolic saponification in the central nervous system to generate the corresponding carboxylic acid form that selectively binds to the excitatory amino acid transporter 2 (EAAT2) protein. PROCEDURES AND METHODS A multi-step high molar activity tracer radiosynthesis was devised to produce doses. Eight healthy human participants (four male and four female), aged 56-75, received a bolus intravenous injection of [18F]RP-115 (administered activity range: 70.3-355 MBq) prior to a total of 94 min of PET-MR scanning performed as three sequential scanning sessions. Regional tissue volumes of interest were defined, time-integrated activity coefficients (TIAC) were derived, and then estimates of organ and tissue activities and effective doses (whole body) were calculated, with two versions of OLINDA software (1.1 and 2.0) that incorporated two tissue weighting factor sets (ICRP-60 and -103), respectively. MAIN FINDINGS Tracer was routinely produced in good radiochemical yields and as suitable high molar activity doses for injection. The [18F]RP-115 injections and PET-MR scans were well-tolerated and no adverse events were reported (≤48 h). Radioactivity was widely biodistributed with good organ uptake. TIACs and estimated radiation organ doses were determined, for which a few statistically significant estimated organ dose differences between males and females were noted. The kidneys were identified as the critical target organ. PRINCIPAL CONCLUSIONS Injection of [18F]RP-115 was considered safe. The estimated kidney radiation doses relative to administered radioactivity identified a more optimal human [18F]RP-115 tracer injected amount of <211 MBq. This more optimal [18F]RP-115 tracer injected activity definition is similar to the amounts used for other established [18F]labeled clinical PET tracers such as [18F]FDG, and it will be used in future RP-115 clinical PET imaging studies.
Collapse
Affiliation(s)
- Chih-Kai Chao
- Rio Pharmaceuticals, Inc., 18 Elsie St., San Francisco, CA 94110, USA.
| | - Joseph Blecha
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - Ilona Polvoy
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA
| | - Ryan Michael Nillo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - John R Forsayeth
- Rio Pharmaceuticals, Inc., 18 Elsie St., San Francisco, CA 94110, USA.
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - John M Gerdes
- Rio Pharmaceuticals, Inc., 18 Elsie St., San Francisco, CA 94110, USA.
| |
Collapse
|
29
|
Brandt JS, Oyelese Y. Foreword: Clinically Focused Insights on the Placenta and Umbilical Cord: An Evidence-based Symposium. Clin Obstet Gynecol 2025; 68:68-71. [PMID: 39648353 DOI: 10.1097/grf.0000000000000920] [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: 12/10/2024]
Abstract
In this symposium, we introduce a collection of reviews that delve into the diverse clinically relevant aspects of the placenta and umbilical cord. The symposium addresses placenta previa and abruption; pathology, genetics, and imaging of the placenta; infections of the placenta; and ischemic placental disease. The umbilical cord's essential function as a fetal lifeline is explored, with an emphasis on the clinical repercussions of its dysfunction, including vasa previa and other umbilical cord abnormalities. This curated collection of reviews, which synthesizes the placenta's and umbilical cord's fundamental role in maternal-fetal health, underscores the clinical importance of these structures in pregnancy.
Collapse
Affiliation(s)
- Justin S Brandt
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, NYU Grossman School of Medicine
- NYU Langone Health, New York, NY
| | - Yinka Oyelese
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center
- Harvard Medical School
- Fetal Surgery and Care Center, Department of Surgery, Boston Children's Hospital, Boston, MA
| |
Collapse
|
30
|
Lohrberg M, Mortensen LS, Thomas C, Fries F, van der Meer F, Götz A, Landt C, Rhee HJ, Rhee J, Gómez-Varela D, Schmidt M, Möbius W, Ruhwedel T, Pardo LA, Remling L, Kramann N, Wrzos C, Bahn E, Stadelmann C, Barrantes-Freer A. Astroglial modulation of synaptic function in the non-demyelinated cerebellar cortex is dependent on MyD88 signaling in a model of toxic demyelination. J Neuroinflammation 2025; 22:47. [PMID: 39988657 PMCID: PMC11849172 DOI: 10.1186/s12974-025-03368-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/03/2025] [Indexed: 02/25/2025] Open
Abstract
Progressive neurological decline in multiple sclerosis is associated with axonal loss and synaptic dysfunction in the non-demyelinated normal appearing gray matter (NAGM) and prominently in the cerebellum. In contrast to early disease stages, where synaptic and neuro-axonal pathology correlates with the extent of T cell infiltration, a prominent role of the innate immune system has been proposed for progressive MS. However, the specific contribution of microglia and astrocytes to synaptic cerebellar pathology in the NAGM- independent of an adaptive T cell response - remains largely unexplored. In the present study, we quantified synaptic changes in the cerebellar NAGM distant from demyelinated lesions in a mouse model of toxic demyelination. Proteomic analysis of the cerebellar cortex revealed differential regulation of synaptic and glutamate transport proteins in the absence of evident structural synaptic pathology or local gray matter demyelination. At the functional level, synaptic changes manifested as a reduction in frequency-dependent facilitation at the parallel fiber- Purkinje cell synapse. Further, deficiency of MyD88, an adaptor protein of the innate immune response, associated with a functional recovery in facilitation, reduced changes in the differential expression of synaptic and glutamate transport proteins, and reduced transcription levels of inflammatory cytokines. Nevertheless, the characteristics of demyelinating lesions and their associated cellular response were similar to wild type animals. Our work brings forward an experimental paradigm mimicking the diffuse synaptic pathology independent of demyelination in late stage MS and highlights the complex regulation of synaptic pathology in the cerebellar NAGM. Moreover, our findings suggest a role of astrocytes, in particular Bergmann glia, as key cellular determinants of cerebellar synaptic dysfunction.
Collapse
Affiliation(s)
- Melanie Lohrberg
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Campus Institute Data Science, Göttingen, Germany
| | - Lena Sünke Mortensen
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
- Paul-Flechsig-Institute of Neuropathology, University Medical Center Leipzig, Leipzig, Germany
| | - Carolina Thomas
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Paul-Flechsig-Institute of Neuropathology, University Medical Center Leipzig, Leipzig, Germany
| | - Franziska Fries
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Alexander Götz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Carolin Landt
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Hong Jun Rhee
- Department of Molecular Neurobiology, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - David Gómez-Varela
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Manuela Schmidt
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Wiebke Möbius
- Department of Neurogenetics, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Network of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Luis A Pardo
- Oncophysiology Group, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Linus Remling
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Nadine Kramann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Claudia Wrzos
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Erik Bahn
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Network of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| | - Alonso Barrantes-Freer
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.
- Paul-Flechsig-Institute of Neuropathology, University Medical Center Leipzig, Leipzig, Germany.
| |
Collapse
|
31
|
Scacco S, Acquaviva S, França Vieira e Silva F, Zhang JH, Lo Muzio L, Corso G, Caponio VCA, Reveglia P, Lecce L, Bizzoca ME, Sherchan P, Cantore S, Ballini A. Bioactivity and Neuroprotective Effects of Extra Virgin Olive Oil in a Mouse Model of Cerebral Ischemia: An In Vitro and In Vivo Study. Int J Mol Sci 2025; 26:1771. [PMID: 40004234 PMCID: PMC11855186 DOI: 10.3390/ijms26041771] [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: 12/31/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Cerebral ischemia is a pathological condition characterized by complete blood and oxygen supply deprivation to neuronal tissue. The ischemic brain compensates for the rapid decline in ATP levels by increasing the anaerobic glycolysis rate, which leads to lactate accumulation and subsequent acidosis. Astrocytes play a critical role in regulating cerebral energy metabolism. Mitochondria are significant targets in hypoxia-ischemia injury, and disruptions in mitochondrial homeostasis and cellular energetics worsen outcomes, especially in the elderly. Elevated levels of n-3 polyunsaturated fatty acids (PUFAs) protect the adult and neonatal brain from ischemic damage by suppressing inflammation, countering oxidative stress, supporting neurovascular unit reconstruction, and promoting oligodendrogenesis. This study examines extra virgin olive oil (EVOO) treatment on TNC WT and TNC M23 cells, focusing on oxygen consumption and reactive oxygen species (ROS) production. This study investigates the effects of different durations of middle cerebral artery occlusion (MCAo) and EVOO administration on cerebral infarct volume, neurological scores, mitochondrial function, and cell viability. Cerebral infarct volume increased with longer ischemia times, while EVOO treatment (0.5 mg/kg/day) significantly reduced infarction across all MCAo durations. The oxygen consumption assays demonstrate EVOO's dose-dependent stimulation of mitochondrial respiration in astrocytes, particularly at lower concentrations. Furthermore, EVOO-treated cells reduce ROS production during hypoxia, improve cell viability under ischemic stress, and enhance ATP production in ischemic conditions, underscoring EVOO's neuroprotective potential.
Collapse
Affiliation(s)
- Salvatore Scacco
- Clinical Biochemistry Unit, Department of Translational Biomedicine and Neuroscience-DiBraiN, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.S.); (S.A.)
| | - Silvia Acquaviva
- Clinical Biochemistry Unit, Department of Translational Biomedicine and Neuroscience-DiBraiN, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.S.); (S.A.)
| | - Fábio França Vieira e Silva
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio, 7, 80138 Naples, Italy;
| | - John H. Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA;
- Department of Anesthesiology, Loma Linda University, Loma Linda, CA 92350, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (G.C.); (V.C.A.C.); (P.R.); (L.L.); (M.E.B.); (A.B.)
| | - Gaetano Corso
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (G.C.); (V.C.A.C.); (P.R.); (L.L.); (M.E.B.); (A.B.)
| | - Vito Carlo Alberto Caponio
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (G.C.); (V.C.A.C.); (P.R.); (L.L.); (M.E.B.); (A.B.)
| | - Pierluigi Reveglia
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (G.C.); (V.C.A.C.); (P.R.); (L.L.); (M.E.B.); (A.B.)
| | - Lucia Lecce
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (G.C.); (V.C.A.C.); (P.R.); (L.L.); (M.E.B.); (A.B.)
| | - Maria Eleonora Bizzoca
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (G.C.); (V.C.A.C.); (P.R.); (L.L.); (M.E.B.); (A.B.)
| | - Prativa Sherchan
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Stefania Cantore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio, 7, 80138 Naples, Italy;
| | - Andrea Ballini
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (L.L.M.); (G.C.); (V.C.A.C.); (P.R.); (L.L.); (M.E.B.); (A.B.)
| |
Collapse
|
32
|
Jang YJ, Kang SJ, Park HS, Lee DH, Kim JH, Kim JE, Kim DI, Chung CH, Yoon JK, Bhang SH. Drug delivery strategies with lipid-based nanoparticles for Alzheimer's disease treatment. J Nanobiotechnology 2025; 23:99. [PMID: 39930497 PMCID: PMC11809104 DOI: 10.1186/s12951-025-03109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/11/2025] [Indexed: 02/14/2025] Open
Abstract
Alzheimer's disease (AD) is a distinctive form of dementia characterized by age-related cognitive decline and memory impairment. A key hallmark of AD is the irreversible overaccumulation of beta-amyloid (Aβ) in the brain, associated with neuroinflammation and neuronal death. Although Aβ clearance and immunoregulation have been the major therapeutic strategies for AD, highly selective transport across the blood-brain barrier (BBB) negatively affects the delivery efficacy of the drugs without the ability to cross the BBB. In this review, we discuss the potential of lipid-based nanoparticles (LBNs) as promising vehicles for drug delivery in AD treatment. LBNs, composed of phospholipid mono- or bilayer, have attracted attention due to their exceptional cellular penetration capabilities and drug loading capabilities, which also facilitate cargo transcytosis across the BBB. Recent advances in the development and engineering of LBNs overcome the existing limitations of the current clinical approaches for AD treatment by addressing off-target effects and low therapeutic efficacy. Here, we review the transport pathways across the BBB, as well as various types of LBNs for AD therapy, including exosomes, liposomes, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs), to elucidate their distinctive properties, preparation methodologies, and therapeutic efficacy, thereby offering innovative avenues for novel drug development for clinical translation in AD therapy.
Collapse
Affiliation(s)
- Young-Ju Jang
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Seong-Jun Kang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea
| | - Hyun Su Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Jae Hoon Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea
| | - Ju-El Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Chan-Hwa Chung
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
33
|
Rosito M, Maqbool J, Reccagni A, Mangano M, D'Andrea T, Rinaldi A, Peruzzi G, Silvestri B, Rosa A, Trettel F, D'Alessandro G, Catalano M, Fucile S, Limatola C. Ketogenic diet induces an inflammatory reactive astrocytes phenotype reducing glioma growth. Cell Mol Life Sci 2025; 82:73. [PMID: 39921723 PMCID: PMC11807044 DOI: 10.1007/s00018-025-05600-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/23/2024] [Accepted: 01/16/2025] [Indexed: 02/10/2025]
Abstract
The use of a ketogenic diet (KD) in glioma is currently tested as an adjuvant treatment in standard chemotherapy regimens. The metabolic shift induced by the KD leads to the generation of ketone bodies that can influence glioma cells and the surrounding microenvironment, but the mechanisms have not yet been fully elucidated. Here, we investigated the potential involvement of glial cells as mediators of the KD-induced effects on tumor growth and survival rate in glioma-bearing mice. Specifically, we describe that exposing glioma-bearing mice to a KD or to β-hydroxybutyrate (β-HB), one of the main KD metabolic products, reduced glioma growth in vivo, induced a pro-inflammatory phenotype in astrocytes and increased functional glutamate transporters. Moreover, we described increased intracellular basal Ca2+ levels in GL261 glioma cells treated with β-HB or co-cultured with astrocytes. These data suggest that pro-inflammatory astrocytes triggered by β-HB can be beneficial in counteracting glioma proliferation and neuronal excitotoxicity, thus protecting brain parenchyma.
Collapse
Affiliation(s)
- Maria Rosito
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy.
- Center for Life Nanoscience & Neuroscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy.
| | - Javeria Maqbool
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Alice Reccagni
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Micol Mangano
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | | | - Arianna Rinaldi
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nanoscience & Neuroscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | - Beatrice Silvestri
- Center for Life Nanoscience & Neuroscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Alessandro Rosa
- Center for Life Nanoscience & Neuroscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Flavia Trettel
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
- IRCCS Neuromed, Pozzilli, IS, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Sergio Fucile
- Department of Physiology and Pharmacology, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy
- IRCCS Neuromed, Pozzilli, IS, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, IS, Italy.
- Department of Physiology and Pharmacology, Laboratory Affiliated to Institute Pasteur Italia, Sapienza University, P.Le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
34
|
Wood OWG, Yeung JHY, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, Kwakowsky A. Upregulated excitatory amino acid transporter 1 (EAAT1) expression in the human medial temporal lobe in Alzheimer's disease. Neuroscience 2025; 566:87-96. [PMID: 39694320 DOI: 10.1016/j.neuroscience.2024.12.027] [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: 09/09/2024] [Revised: 11/30/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Alzheimer's disease (AD) is a growing health problem worldwide, particularly in the developed world due to an ageing population. Glutamate excitotoxicity plays a major role in the pathophysiology of AD, and glutamate re-uptake is controlled by excitatory amino acid transporters (EAATs). The EAAT2 isoform is the predominant transporter involved in glutamate reuptake, therefore EAAT1 has not been the focus of AD research. We investigated the layer-specific expression of EAAT1 in human medial temporal lobe regions such as the hippocampus, subiculum, entorhinal cortex and superior temporal gyrus, using fluorescent immunohistochemistry and laser scanning confocal microscopy in human post-mortem tissue. We observed low EAAT1 immunoreactivity in control cases, but upregulated labeling in AD across several brain regions of the medial temporal lobe. Significantly higher integrated density in AD cases was observed in the str. oriens and str. radiatum of the CA2 region, the str. pyramidale of CA3, and the str. moleculare and str. granulosum of the DG. Labeling of EAAT1 appeared astrocytic in nature, showing close association with astrocytic processes in AD cases. We also report that a higher EAAT1 density was positively correlated with the age of AD cases, but this relationship was not observed in control cases. Overall, our results indicate an upregulation of EAAT1 across several hippocampal subregions and layers in AD cases, indicating a potential physiological role for this transporter that needs further investigation.
Collapse
Affiliation(s)
- Oliver W G Wood
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Jason H Y Yeung
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Clinton Turner
- Department of Anatomical Pathology, LabPLUS, Auckland City Hospital, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand; Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Ireland.
| |
Collapse
|
35
|
Nayak U, Manikkath J, Arora D, Mudgal J. Impact of neuroinflammation on brain glutamate and dopamine signalling in schizophrenia: an update. Metab Brain Dis 2025; 40:119. [PMID: 39907868 PMCID: PMC11799129 DOI: 10.1007/s11011-025-01548-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025]
Abstract
Schizophrenia is one of the most severe and chronic psychiatric disorders. Over the years, numerous treatment options have been introduced for schizophrenia. Although they are relatively successful in managing the positive symptoms of schizophrenia, most of the current treatments have a negligible effect on the negative and cognitive symptoms. Thus, none of them could prevent the relapse of psychotic episodes. Among the numerous hypotheses explaining the development and progression of schizophrenia, the cytokine hypothesis explains the role of inflammatory markers as a significant culprit in the development of schizophrenia. Elevated cytokines are reported in animal models and schizophrenic patients. The cytokine hypothesis is based on how increased inflammatory markers can cause changes in the dopaminergic, glutamate, and tryptophan metabolism pathways, like that observed in schizophrenic patients. Reasons, such as autoimmune disease, maternal immune activation, infection, etc., can pave the way for the development of schizophrenia and are associated with the negative, positive and cognitive symptoms of schizophrenia. Thus, there is a need to focus on the significance of anti-inflammatory drugs against these symptoms. The development of new treatment strategies in the management of schizophrenia can provide better therapeutic outcomes in terms of the severity of symptoms and treatment of drug-resistant schizophrenia. This review attempts to explain the association between elevated inflammatory markers and various neurotransmitters, and the possible use of medications like nonsteroidal anti-inflammatory drugs, monoclonal antibodies, statins, and estrogens as adjuvant therapy. Over the years, these hypotheses have been the basis for drug discovery for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Usha Nayak
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Jyothsna Manikkath
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Devinder Arora
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
36
|
Garcia JP, Armbruster M, Sommer M, Nunez-Beringer A, Dulla CG. Glutamate uptake is transiently compromised in the perilesional cortex following controlled cortical impact. Cereb Cortex 2025; 35:bhaf031. [PMID: 40007051 DOI: 10.1093/cercor/bhaf031] [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: 08/28/2024] [Revised: 12/03/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Glutamate, the primary excitatory neurotransmitter in the central nervous system (CNS), is regulated by the excitatory amino acid transporters glutamate transporter 1 (GLT-1) and glutamate aspartate transporter (GLAST). Following traumatic brain injury, extracellular glutamate levels increase, contributing to excitotoxicity, circuit dysfunction, and morbidity. Increased neuronal glutamate release and compromised astrocyte-mediated uptake contribute to elevated glutamate, but the mechanistic and spatiotemporal underpinnings of these changes are not well established. Using the controlled cortical impact model of TBI and iGluSnFR glutamate imaging, we quantified extracellular glutamate dynamics after injury. Three days postinjury, glutamate release was increased, and glutamate uptake and GLT-1 expression were reduced. Seven and 14 days postinjury, glutamate dynamics were comparable between sham and controlled cortical impact animals. Changes in peak glutamate response were unique to specific cortical layers and proximity to injury. This was likely driven by increases in glutamate release, which was spatially heterogeneous, rather than reduced uptake, which was spatially uniform. The astrocyte K+ channel, Kir4.1, regulates activity-dependent slowing of glutamate uptake. Surprisingly, Kir4.1 was unchanged after controlled cortical impact and accordingly, activity-dependent slowing of glutamate uptake was unaltered. This dynamic glutamate dysregulation after traumatic brain injury underscores a brief period in which disrupted glutamate uptake may contribute to dysfunction and highlights a potential therapeutic window to restore glutamate homeostasis.
Collapse
Affiliation(s)
- Jacqueline P Garcia
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
- Cellular, Molecular, and Developmental Biology Program, Tufts University School of Medicine, Boston, MA, United States
| | - Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Mary Sommer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Aliana Nunez-Beringer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
37
|
Rodríguez-Campuzano AG, Castelán F, Hernández-Kelly LC, Felder-Schmittbuhl MP, Ortega A. Yin Yang 1: Function, Mechanisms, and Glia. Neurochem Res 2025; 50:96. [PMID: 39904836 PMCID: PMC11794380 DOI: 10.1007/s11064-025-04345-7] [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: 10/31/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Yin Yang 1 is a ubiquitously expressed transcription factor that has been extensively studied given its particular dual transcriptional regulation. Yin Yang 1 is involved in various cellular processes like cell cycle progression, cell differentiation, DNA repair, cell survival and apoptosis among others. Its malfunction or alteration leads to disease and even to malignant transformation. This transcription factor is essential for the proper central nervous system development and function. The activity of Yin Yang 1 depends on its interacting partners, promoter environment and chromatin structure, however, its mechanistic activity is not completely understood. In this review, we briefly discuss the Yin Yang 1 structure, post-translational modifications, interactions, mechanistic functions and its participation in neurodevelopment. We also discuss its expression and critical involvement in the physiology and physiopathology of glial cells, summarizing the contribution of Yin Yang 1 on different aspects of cellular function.
Collapse
Affiliation(s)
- Ada G Rodríguez-Campuzano
- Departamento de Biología Celular y Fisiología, Unidad Foránea Tlaxcala, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlaxcala, Tlaxcala, Mexico
| | - Francisco Castelán
- Departamento de Biología Celular y Fisiología, Unidad Foránea Tlaxcala, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlaxcala, Tlaxcala, Mexico
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacantenco, G.A. Madero, 07360, Ciudad de Mexico, Mexico
| | - Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), Université de Strasbourg, Strasbourg, France
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacantenco, G.A. Madero, 07360, Ciudad de Mexico, Mexico.
| |
Collapse
|
38
|
Zhang Z, Huang W, Zhang X, Wang Z, Xie M, Xie B, Wang Y, Chen X, Xiang AP, Xiang Q. Human iPSC-derived mesenchymal stem cells relieve high blood pressure in spontaneously hypertensive rats via splenic nerve activated choline acetyltransferase-positive cells. SCIENCE CHINA. LIFE SCIENCES 2025; 68:502-514. [PMID: 39428428 DOI: 10.1007/s11427-023-2675-2] [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: 03/08/2024] [Accepted: 06/28/2024] [Indexed: 10/22/2024]
Abstract
Despite substantial advancements in modern medicine, the management of hypertension remains a major challenge. Stem cell-based therapies have recently demonstrated remarkable efficacy in treating cardiovascular diseases, including hypertension. However, the antihypertensive mechanism of mesenchymal stem cells (MSCs) has not been extensively explored. This study aimed to investigate the role of injected MSCs in regulating blood pressure homeostasis. Our previous study demonstrated that human induced pluripotent stem cell-derived mesenchymal stem cells (hiPSC-MSCs) are functional and homogeneous sources for MSC-based therapy. After the injection of hiPSC-MSCs, a significant reduction in blood pressure and end target organ inflammation were observed in spontaneously hypertensive rats (SHRs). Cell tracking assays demonstrated that the injected hiPSC-MSCs accumulated in the spleens of the SHRs. The injected hiPSC-MSCs accumulated adjacent to the splenic nerve, potentially contributing to the antihypertensive effects. Furthermore, the hiPSC-MSCs released abundant glutamate, which acts as a neuromodulator to activate the splenic sympathetic nerve. After inhibition of glutamate synthesis by siRNA, the ability of hiPSC-MSCs to activate sympathetic nerves was significantly diminished. In addition, the antihypertensive effects of hiPSC-MSCs were eliminated after splenic nerve denervation (SND), underscoring the critical role of the splenic nerve. Moreover, activation of the splenic nerve resulted in increased release of norepinephrine (NE), which increased the number of choline acetyltransferase-positive (ChAT+) cells in the spleen and peripheral blood. Consequently, the acetylcholine (ACh) produced by elevated ChAT+ cells could act as a vasodilator, lowering blood pressure and mitigating inflammation in end target organs. In summary, our findings indicate that hiPSC-MSCs effectively lower blood pressure in hypertension by influencing the splenic nerves and regulating ChAT+ cells. The connection between blood pressure regulation and the splenic nerve may offer new insights into the treatment of hypertension.
Collapse
Affiliation(s)
- Zhen Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhecun Wang
- Department of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510062, China
| | - Manting Xie
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Bingbing Xie
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yiling Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Qiuling Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
39
|
Huang Q, Lee HH, Volpe B, Zhang Q, Xue C, Liu BC, Abuhasan YR, Li L, Yang JS, Egholm J, Gutierrez-Vazquez C, Li A, Lee A, Tang S, Wong CW, Liu T, Huang Y, Ramos RL, Stout RF, El Ouaamari A, Quintana FJ, Lowell BB, Kahn CR, Pothos EN, Cai W. Deletion of murine astrocytic vesicular nucleotide transporter increases anxiety and depressive-like behavior and attenuates motivation for reward. Mol Psychiatry 2025; 30:506-520. [PMID: 39122778 PMCID: PMC11750621 DOI: 10.1038/s41380-024-02692-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Astrocytes are multi-functional glial cells in the central nervous system that play critical roles in modulation of metabolism, extracellular ion and neurotransmitter levels, and synaptic plasticity. Astrocyte-derived signaling molecules mediate many of these modulatory functions of astrocytes, including vesicular release of ATP. In the present study, we used a unique genetic mouse model to investigate the functional significance of astrocytic exocytosis of ATP. Using primary cultured astrocytes, we show that loss of vesicular nucleotide transporter (Vnut), a primary transporter responsible for loading cytosolic ATP into the secretory vesicles, dramatically reduces ATP loading into secretory lysosomes and ATP release, without any change in the molecular machinery of exocytosis or total intracellular ATP content. Deletion of astrocytic Vnut in adult mice leads to increased anxiety, depressive-like behaviors, and decreased motivation for reward, especially in females, without significant impact on food intake, systemic glucose metabolism, cognition, or sociability. These behavioral alterations are associated with significant decreases in the basal extracellular dopamine levels in the nucleus accumbens. Likewise, ex vivo brain slices from these mice show a strong trend toward a reduction in evoked dopamine release in the nucleus accumbens. Mechanistically, the reduced dopamine signaling we observed is likely due to an increased expression of monoamine oxidases. Together, these data demonstrate a key modulatory role of astrocytic exocytosis of ATP in anxiety, depressive-like behavior, and motivation for reward, by regulating the mesolimbic dopamine circuitry.
Collapse
Affiliation(s)
- Qian Huang
- Department of Molecular and Cellular Biochemistry, the Barnstable Brown Diabetes and Obesity Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Hiu Ham Lee
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Bryan Volpe
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Qingchen Zhang
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Chang Xue
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Brian C Liu
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Yahia R Abuhasan
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Lingyun Li
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Jeremy S Yang
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Julie Egholm
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Cristina Gutierrez-Vazquez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Allen Li
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Alyssa Lee
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Sharon Tang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Chun Wa Wong
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Tiemin Liu
- Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Yuan Huang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Raddy L Ramos
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Randy F Stout
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | | | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Emmanuel N Pothos
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Weikang Cai
- Department of Molecular and Cellular Biochemistry, the Barnstable Brown Diabetes and Obesity Center, University of Kentucky College of Medicine, Lexington, KY, USA.
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA.
| |
Collapse
|
40
|
Zhu HH, Song XK, Zhang JL, Zhang JL, Rong PJ, Wang JY. Involvement of the astroglial glutamate-glutamine cycle in the analgesic effects of electroacupuncture in a rat model of chronic neuropathic pain. Acupunct Med 2025; 43:14-25. [PMID: 39912444 DOI: 10.1177/09645284241309952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
OBJECTIVE Our previous study found that astrocytes are involved in cumulative analgesia; however, the underlying mechanism remains unclear. The aim of this study was to further explore the potential role of astrocytes in the effects of electroacupuncture (EA) on neuropathic pain by focusing on the glutamate-glutamine cycle. METHODS 69 male Sprague-Dawley (SD) rats were randomly divided into a normal control group, untreated chronic constriction injury (CCI) model group and EA-treated model (CCI + EA) group. EA was applied bilaterally at ST36 and GB34. Pain thresholds were assessed using behavioral tests and thermal stimuli. We examined the co-expression of glutamate/aspartate transporter (GLAST) via immunofluorescence and measured the expression levels of GLAST, glutamate transporter (GLT)-1 and glutamine synthetase (GS) using Western blotting and polymerase chain reaction (PCR). Glutamate (Glu) and gamma-aminobutyric acid (GABA) levels were detected by high-performance liquid chromatography (HPLC). To validate the impact of GLAST/GLT-1 in the analgesic effect of EA, an additional 30 SD male rats were divided into groups receiving intrathecal saline, GLAST antagonist or GLT-1 antagonist alongside EA. RESULTS Post-CCI, pain thresholds were decreased, GLAST expression was diminished, and spinal Glu levels were increased. EA treatment reversed these effects, improved pain thresholds and GLAST/GLT-1 expression in astrocytes, and reduced Glu levels. Antagonist administration negated the analgesic effects of EA. CONCLUSION Repeated EA administration inhibited CCI-induced chronic neuropathic pain in rats, corresponding to a reversal of decreased expression of GLAST and GLT-1, which may have accelerated the clearance of Glu and thereby reduced its concentration. Regulation of the astroglial glutamate-glutamine cycle is a potential target of EA.
Collapse
Affiliation(s)
- Hao-Han Zhu
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xing-Ke Song
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian-Liang Zhang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jin-Ling Zhang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Pei-Jing Rong
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun-Ying Wang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
41
|
Mohammad ZB, Yudin SCY, Goldberg BJ, Serra KL, Klegeris A. Exploring neuroglial signaling: diversity of molecules implicated in microglia-to-astrocyte neuroimmune communication. Rev Neurosci 2025; 36:91-117. [PMID: 39240134 PMCID: PMC11717358 DOI: 10.1515/revneuro-2024-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024]
Abstract
Effective communication between different cell types is essential for brain health, and dysregulation of this process leads to neuropathologies. Brain glial cells, including microglia and astrocytes, orchestrate immune defense and neuroimmune responses under pathological conditions during which interglial communication is indispensable. Our appreciation of the complexity of these processes is rapidly increasing due to recent advances in molecular biology techniques, which have identified numerous phenotypic states of both microglia and astrocytes. This review focuses on microglia-to-astrocyte communication facilitated by secreted neuroimmune modulators. The combinations of interleukin (IL)-1α, tumor necrosis factor (TNF), plus complement component C1q as well as IL-1β plus TNF are already well-established microglia-derived stimuli that induce reactive phenotypes in astrocytes. However, given the large number of inflammatory mediators secreted by microglia and the rapidly increasing number of distinct functional states recognized in astrocytes, it can be hypothesized that many more intercellular signaling molecules exist. This review identifies the following group of cytokines and gliotransmitters that, while not established as interglial mediators yet, are known to be released by microglia and elicit functional responses in astrocytes: IL-10, IL-12, IL-18, transforming growth factor (TGF)-β, interferon (IFN)-γ, C-C motif chemokine ligand (CCL)5, adenosine triphosphate (ATP), l-glutamate, and prostaglandin E2 (PGE2). The review of molecular mechanisms engaged by these mediators reveals complex, partially overlapping signaling pathways implicated in numerous neuropathologies. Additionally, lack of human-specific studies is identified as a significant knowledge gap. Further research on microglia-to-astrocyte communication is warranted, as it could discover novel interglial signaling-targeted therapies for diverse neurological disorders.
Collapse
Affiliation(s)
- Zainab B. Mohammad
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Samantha C. Y. Yudin
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Benjamin J. Goldberg
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Kursti L. Serra
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Andis Klegeris
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| |
Collapse
|
42
|
Pietrobon D, Brennan KC. Mechanisms underlying CSD initiation implicated by genetic mouse models of migraine. J Headache Pain 2025; 26:17. [PMID: 39871148 PMCID: PMC11773941 DOI: 10.1186/s10194-025-01948-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
A key unanswered question in migraine neurobiology concerns the mechanisms that make the brain of migraineurs susceptible to cortical spreading depression (CSD, a spreading depolarization that underlies migraine aura and may trigger the migraine pain mechanisms). Important insights into this question can be obtained by studying the mechanisms of facilitation of CSD initiation in genetic mouse models of the disease. These models, all generated from families with hereditary migraine, allow the investigation of the functional consequences of disease-causing mutations at the molecular, cellular, synaptic and neural circuit levels. In this review, after describing the available genetic mouse models of migraine, which all share increased susceptibility to experimentally induced CSD, we will discuss the functional alterations in their cerebral cortex and the mechanisms underlying the facilitation of CSD initiation in their cortex, as well as the insights that these mechanisms may give into the mechanisms of initiation of spontaneous CSDs in migraine.
Collapse
Affiliation(s)
- Daniela Pietrobon
- Department of Biomedical Sciences and Padova Neuroscience Center, University of Padova, Via Ugo Bassi 58, 35131, Padua, Italy.
| | - K C Brennan
- Department of Neurology, University of Utah, 383 Colorow Drive, Salt Lake City, UT, 84108, USA.
| |
Collapse
|
43
|
He J, Hewett SJ. Nrf2 Regulates Basal Glutathione Production in Astrocytes. Int J Mol Sci 2025; 26:687. [PMID: 39859401 PMCID: PMC11765531 DOI: 10.3390/ijms26020687] [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: 11/11/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Astrocytes produce and export glutathione (GSH), an important thiol antioxidant essential for protecting neural cells from oxidative stress and maintaining optimal brain health. While it has been established that oxidative stress increases GSH production in astrocytes, with Nrf2 acting as a critical transcription factor regulating key components of the GSH synthetic pathway, the role of Nrf2 in controlling constitutive GSH synthetic and release mechanisms remains incompletely investigated. Our data show that naïve primary mouse astrocytes cultured from the cerebral cortices of Nrf2 knockout (Nrf2-/-) pups have significantly less intracellular and extracellular GSH levels when compared to astrocytes cultured from Nrf2 wild-type (Nrf2+/+) pups. Key components of the GSH synthetic pathway, including xCT (the substrate-specific light chain of the substrate-importing transporter, system xc-), glutamate-cysteine ligase [catalytic (GCLc) and modifying (GCLm) subunits], were affected. To wit: qRT-PCR analysis demonstrates that naïve Nrf2-/- astrocytes have significantly lower basal mRNA levels of xCT and both GCL subunits compared to naïve Nrf2+/+ astrocytes. No change in mRNA levels of glutathione synthetase (GS) or the GSH exporting transporter, Mrp1, was found. Western blot analysis reveals reduced protein levels of both subunits of GCL, while (seleno)cystine uptake into Nrf2-/- astrocytes was reduced compared to Nrf2+/+ astrocytes, confirming decreased system xc- activity. These findings suggest that Nrf2 regulates the basal production of GSH in astrocytes through constitutive transcriptional regulation of GCL and xCT.
Collapse
Affiliation(s)
| | - Sandra J. Hewett
- Program in Neuroscience, Department of Biology, Syracuse University, Syracuse, NY 13210, USA;
| |
Collapse
|
44
|
Martinez-Lozada Z, Guillem AM, Song I, Gonzalez MV, Takano H, Parikh E, Rothstein JD, Putt ME, Robinson MB. Identification of a Subpopulation of Astrocyte Progenitor Cells in the Neonatal Subventricular Zone: Evidence that Migration is Regulated by Glutamate Signaling. Neurochem Res 2025; 50:77. [PMID: 39789409 PMCID: PMC11717811 DOI: 10.1007/s11064-024-04326-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
In mice engineered to express enhanced green fluorescent protein (eGFP) under the control of the entire glutamate transporter 1 (GLT1) gene, eGFP is found in all 'adult' cortical astrocytes. However, when 8.3 kilobases of the human GLT1/EAAT2 promoter is used to control expression of tdTomato (tdT), tdT is only found in a subpopulation of these eGFP-expressing astrocytes. The eGFP mice have been used to define mechanisms of transcriptional regulation using astrocytes cultured from cortex of 1-3 day old mice. Using the same cultures, we were never able to induce tdT+ expression. We hypothesized that these cells might not have migrated into the cortex by this age. In this study, we characterized the ontogeny of tdT+ cells, performed single-cell RNA sequencing (scRNA-seq), and tracked their migration in organotypic slice cultures. At postnatal day (PND) 1, tdT+ cells were observed in the subventricular zone and striatum but not in the cortex, and they did not express eGFP. At PND7, tdT+ cells begin to appear in the cortex with their numbers increasing with age. At PND1, scRNA-seq demonstrates that the tdT+ cells are molecularly heterogeneous, with a subpopulation expressing astrocytic markers, subsequently validated with immunofluorescence. In organotypic slices, tdT+ cells migrate into the cortex, and after 7 days they express GLT1, NF1A, and eGFP. An ionotropic glutamate receptor (iGluR) antagonist reduced by 50% the distance tdT+ cells migrate from the subventricular zone into the cortex. The pan-glutamate transport inhibitor, TFB-TBOA, increased, by sixfold, the number of tdT+ cells in the cortex. In conclusion, although tdT is expressed by non-glial cells at PND1, it is also expressed by glial progenitors that migrate into the cortex postnatally. Using this fluorescent labeling, we provide novel evidence that glutamate signaling contributes to the control of glial precursor migration.
Collapse
Affiliation(s)
- Zila Martinez-Lozada
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA.
- Department of Neuroscience, College of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL, 33328, USA.
| | - Alain M Guillem
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA
| | - Isabella Song
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA
| | - Michael V Gonzalez
- Center for Cytokine Storm Treatment & Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hajime Takano
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Esha Parikh
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA
| | - Jeffrey D Rothstein
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mary E Putt
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael B Robinson
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA.
| |
Collapse
|
45
|
Wang J, Chen Q, Jiang S, Liu S, Xie Z, Zhang X, Huang H, Zhu S. Regulation of Glutamate Transporter Type 1 by TSA and the Antiepileptic Mechanism of TSA. Neurochem Res 2025; 50:74. [PMID: 39754645 PMCID: PMC11700035 DOI: 10.1007/s11064-024-04317-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025]
Abstract
Epilepsy (EP) is a neurological disorder characterized by abnormal, sudden neuronal discharges. Seizures increase extracellular glutamate levels, causing excitotoxic damage. Glutamate transporter type 1 (GLT-1) and its human homologue excitatory amino acid transporter-2 (EAAT2) clear 95% of extracellular glutamate. Studies on neurodegenerative diseases suggest that trichostatin A (TSA), a broad-spectrum histone deacetylase (HDAC) inhibitor, can increase GLT-1/EAAT2 transcription. However, the precise mechanism by which TSA modulates GLT-1/EAAT2 levels remains unclear. This research demonstrated that TSA increases GLT-1/EAAT2 expression through histone acetylation, exerting substantial antiepileptic effects. Our results identify a promising therapeutic strategy for EP involving the modulation of glutamate transporters to mitigate seizures. Future research should explore the specific mechanisms underlying the effects of TSA and its potential clinical applications. Acute and chronic EP models were induced using kainic acid (KA) to assess the effects of TSA on the seizure threshold and frequency. Electrophysiological recordings of the hippocampus were used to evaluate the impact of TSA on neuronal excitability. RNA-Seq was used to analyse changes in glutamate transporter-related gene expression. Western blot analysis and qRT‒PCR were used to assess the influence of TSA on HDAC expression. To validate the role of GLT-1/EAAT2 in the antiepileptic effects of TSA, the impact of the GLT-1/EAAT2 inhibitor dihydrokainic acid (DHK) on the effects of TSA was assessed. Glutamate release was measured, and microdialysis was used to determine the glutamate content in the cerebrospinal fluid. Finally, metabolomics analysis was used to explore changes in amino acid levels in the hippocampus following TSA treatment to further confirm the antiepileptic potential of TSA. TSA effectively inhibited seizures in both acute and chronic models. It reduced the amplitude of excitatory postsynaptic currents (PSCs) and the frequency of spontaneous excitatory PSCs in the hippocampus without affecting inhibitory PSCs. Transcriptome analysis was used to identify glutamate transmission-related targets and revealed significant upregulation of the GLT-1 and EAAT2 genes in the hippocampus, which was confirmed by qRT‒PCR and Western blotting. Acetylation-induced upregulation of GLT-1/EAAT2 was observed, and inhibition of these transporters by DHK reduced the seizure-mitigating effects of TSA, underscoring the role of GLT-1/EAAT2 in clearing glutamate and its contribution to the observed antiepileptic effects of TSA. Our findings highlight the crucial role of GLT-1/EAAT2 in mediating the impact of TSA on glutamatergic transmission and seizure activity. These insights pave the way for the development of novel therapeutic approaches for EP involving the modulation of glutamate transporters.
Collapse
Affiliation(s)
- Jin Wang
- Huazhong University of Science and Technology, Tongji Medical College, Wuhan, Hubei, 430000, China
- Medical College, Guangxi University of Science and Technology, Liuzhou, Guangxi, 545005, China
- Department of Oncology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, 545005, China
| | - Qinqin Chen
- Huazhong University of Science and Technology, Tongji Medical College, Wuhan, Hubei, 430000, China
- Medical College, Guangxi University of Science and Technology, Liuzhou, Guangxi, 545005, China
- Department of Oncology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, 545005, China
| | - Shihai Jiang
- Medical College, Guangxi University of Science and Technology, Liuzhou, Guangxi, 545005, China
| | - Sisi Liu
- Medical College, Guangxi University of Science and Technology, Liuzhou, Guangxi, 545005, China
| | - Zhengyi Xie
- Medical College, Guangxi University of Science and Technology, Liuzhou, Guangxi, 545005, China
| | - Xiaochen Zhang
- Medical College, Guangxi University of Science and Technology, Liuzhou, Guangxi, 545005, China.
| | - Haixin Huang
- Department of Oncology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, 545005, China.
| | - Suiqiang Zhu
- Huazhong University of Science and Technology, Tongji Medical College, Wuhan, Hubei, 430000, China.
| |
Collapse
|
46
|
Selvaraj S, Weerasinghe L. The Role of Nanotechnology in Understanding the Pathophysiology of Traumatic Brain Injury. Cent Nerv Syst Agents Med Chem 2025; 25:20-38. [PMID: 38676493 DOI: 10.2174/0118715249291999240418112531] [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: 11/09/2023] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/29/2024]
Abstract
Recently, traumatic brain injury (TBI) has been a growing disorder due to frequent brain dysfunction. The Glasgow Coma Scale expresses TBI as classified as having mild, moderate, or severe brain effects, according to the effects on the brain. Brain receptors undergo various modifications in their pathology through chemical synaptic pathways, leading to depression, Alzheimer's, and Parkinson's disease. These brain disorders can be controlled using central receptors such as dopamine, glutamate, and γ-aminobutyric acid, which are clearly explained in this review. Furthermore, there are many complications in TBI's clinical trials and diagnostics, leading to insignificant treatment, causing permanent neuro-damage, physical disability, and even death. Bio-screening and conventional molecular-based therapies are inappropriate due to poor preclinical testing and delayed recovery. Hence, modern nanotechnology utilizing nanopulsed laser therapy and advanced nanoparticle insertion will be suitable for TBI's diagnostics and treatment. In recent days, nanotechnology has an important role in TBI control and provides a higher success rate than conventional therapies. This review highlights the pathophysiology of TBI by comprising the drawbacks of conventional techniques and supports suitable modern alternates for treating TBI.
Collapse
Affiliation(s)
- Saranya Selvaraj
- Department of Chemistry, Faculty of Applied sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Laksiri Weerasinghe
- Department of Chemistry, Faculty of Applied sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| |
Collapse
|
47
|
Sarnat HB, Rao VTS. Neuroglia pathology in genetic and epigenetic disorders of the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:87-99. [PMID: 40148059 DOI: 10.1016/b978-0-443-19102-2.00003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Glial cells are increasingly recognized for their important interactions with both developing and mature neurons, in particular for maintenance of dendritic ramifications and spines, synapses, and neurotransmitter uptake. MicroRNA abnormalities are demonstrated in individual astrocytes with alterations in neurological diseases. Alexander disease is a prototype astrocytic disease because of genetically altered glial fibrillary acidic protein (GFAP) filaments. Other genetic diseases are now recognized as involving glial cells in their pathogenesis: Rett, Fragile-X, Aicardi-Goutières, and Down syndromes, as well as epigenetic effects in the mechanism of fetal alcohol spectrum disorder. Many involve glial production of cytokines and neuroinflammation. Microglia also may contribute. The heat-shock protein α-B-crystallin is expressed in the Rosenthal fibers of Alexander disease, in which the molecular structure of GFAP is altered, in astrocytes secreting neurotoxic cytokines, and focally at or near epileptic foci. Satellite glial cells adherent to neuronal soma are frequent and diagnostically nonspecific but may contribute to neuronal degeneration, especially of hypermetabolic epileptogenic neurons. Glial cells have distorted size and morphology in mTOR malformations. Failure of glial apoptosis in the fetal lamina terminalis is the likely pathogenesis of callosal agenesis and of other cerebral dysgeneses.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Departments of Paediatrics (Neurology), Pathology and Laboratory Medicine (Neuropathology), and Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, AB, Canada; Medical Affairs Laboratory, Argenx US, Inc., Boston, MA, United States.
| | - Vijayaraghava T S Rao
- Departments of Paediatrics (Neurology), Pathology and Laboratory Medicine (Neuropathology), and Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, AB, Canada; Medical Affairs Laboratory, Argenx US, Inc., Boston, MA, United States
| |
Collapse
|
48
|
Zinsmaier AK, Nestler EJ, Dong Y. Astrocytic G Protein-Coupled Receptors in Drug Addiction. ENGINEERING (BEIJING, CHINA) 2025; 44:256-265. [PMID: 40109668 PMCID: PMC11922559 DOI: 10.1016/j.eng.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Understanding the cellular mechanisms of drug addiction remains a key task in current brain research. While neuron-based mechanisms have been extensively explored over the past three decades, recent evidence indicates a critical involvement of astrocytes, the main type of non-neuronal cells in the brain. In response to extracellular stimuli, astrocytes modulate the activity of neurons, synaptic transmission, and neural network properties, collectively influencing brain function. G protein-coupled receptors (GPCRs) expressed on astrocyte surfaces respond to neuron- and environment-derived ligands by activating or inhibiting astrocytic signaling, which in turn regulates adjacent neurons and their circuitry. In this review, we focus on the dopamine D1 receptors (D1R) and metabotropic glutamate receptor 5 (mGLUR5 or GRM5)-two GPCRs that have been critically implicated in the acquisition and maintenance of addiction-related behaviors. Positioned as an introductory-level review, this article briefly discusses astrocyte biology, outlines earlier discoveries about the role of astrocytes in substance-use disorders (SUDs), and provides detailed discussion about astrocytic D1Rs and mGLUR5s in regulating synapse and network functions in the nucleus accumbens (NAc)-a brain region that mediates addiction-related emotional and motivational responses. This review serves as a stepping stone for readers of Engineering to explore links between astrocytic GPCRs and drug addiction and other psychiatric disorders.
Collapse
Affiliation(s)
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
49
|
Lodetti G, Baldin SL, de Farias ACS, de Pieri Pickler K, Teixeira AG, Dondossola ER, Bernardo HT, Maximino C, Rico EP. Repeated exposure to ethanol alters memory acquisition and neurotransmission parameters in zebrafish brain. Pharmacol Biochem Behav 2025; 246:173915. [PMID: 39586362 DOI: 10.1016/j.pbb.2024.173915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
Alcohol is widely consumed worldwide and its abuse can cause cognitive dysfunction, affecting memory and learning due to several neurophysiological changes. An imbalance in several neurotransmitters, including the cholinergic and glutamatergic systems, have been implicated in these effects. Zebrafish are sensitive to alcohol, respond to reward stimuli, and tolerate and exhibit withdrawal behaviors. Therefore, we investigated the effects of repetitive exposure to ethanol (REE) and the NMDA receptor antagonist dizocilpine (MK-801) on memory acquisition and glutamatergic and cholinergic neurotransmission. Memory was assessed using the inhibitory avoidance and object recognition tasks. Brain glutamate levels and the activity of Na+-dependent transporters were evaluated as indexes of glutamatergic activity, while acetylcholinesterase (AChE) and choline acetyltransferase (ChAT), enzyme activity were evaluated as indexes of cholinergic activity. Behavioral assessments showed that REE impaired aversive and spatial memory, an effect that MK-801 mimicked. Glutamate levels, but not transporter activity, were significantly lower in the REE group; similarly, REE increased the activity of AChE, but not ChAT, activity. These findings suggest that intermittent exposure to ethanol leads to impairments in zebrafish memory consolidation, and that these effects could be associated with alterations in parameters related to neurotransmission systems mediated by glutamate and acetylcholine. These results provide a better understanding of the neurophysiological and behavioral changes caused by repetitive alcohol use.
Collapse
Affiliation(s)
- Guilherme Lodetti
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Samira Leila Baldin
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Ana Carolina Salvador de Farias
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Karolyne de Pieri Pickler
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Amanda Gomes Teixeira
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Eduardo Ronconi Dondossola
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Henrique Teza Bernardo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Caio Maximino
- Laboratory of Neurosciences and Behavior, Institute of Health and Biological Studies, Federal University of South and Southeast of Pará, Marabá, Brazil
| | - Eduardo Pacheco Rico
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil.
| |
Collapse
|
50
|
Zang X, Zhang J, Hu J, Mo X, Zheng T, Ji J, Xing J, Chen C, Zhou S. Electroconvulsive therapy combined with esketamine improved depression through PI3K/AKT/GLT-1 pathway. J Affect Disord 2025; 368:282-294. [PMID: 39265873 DOI: 10.1016/j.jad.2024.08.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 09/14/2024]
Abstract
Neuron excitotoxic damage induced by extracellular glutamate accumulation pathologically is one of the main mechanisms of depression. Glutamate transporter-1 (GLT-1) expressed in astrocyte is responsible for glutamate clearance to maintain glutamate balance. Electroconvulsive therapy (ECT) is prevalently recommended for severe depression due to its significant anti-depressant effect. Esketamine could offer advantages of rapid anti-depressant effect and neuron protection. The aim of this study is to investigate the anti-depressant efficacy of esketamine plus ECT, and further to explore the mechanism. Firstly, total 12 patients were randomized into anesthesia with propofol (P) or propofol+esketamine (PK) before ECT. 24-Hamilton Depression Scale (HAMD) was used to evaluate the severity of depression after each ECT. Then, depressive rat model was built using chronic unpredictable mild stress method, and subsequently received infusion of esketamine (5 mg/kg) or saline before ECT treatment (0.5 mA; 100 V) for consecutive 10 days. Tests including sucrose preference test, open field test and forced swimming test were used to evaluate depression-like behaviors. In next experiments, rats were injected with RIL, DHK or LY294002 intracerebroventricularly for continuous 10 days before individual treatment. After the fifth and sixth ECT, PK group displayed lower HAMD score compared to P group. In rat model, we found that esketamine plus ECT could significantly improve depression-like behaviors and decrease glutamate level. Esketamine and ECT could both activate PI3K/Akt/GLT-1 pathway. The GLT-1 agonist RIL made equivalent effect as esketamine plus ECT. Furthermore, after using PI3K/Akt inhibitor LY294002 and GLT-1 inhibitor DHK, esketamine plus ECT could neither improve depression-like symptoms, nor upregulate GLT-1 level. Our present study suggested that esketamine plus ECT could dramatically improve depression symptom. The activation of PI3K/Akt/GLT-1 pathway may be the potential mechanism.
Collapse
Affiliation(s)
- Xiangyang Zang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, PR China
| | - Jingting Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, PR China
| | - Jingping Hu
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, PR China
| | - Xingying Mo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, PR China
| | - Tingwei Zheng
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, PR China
| | - Jiaming Ji
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, PR China
| | - Jibin Xing
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, PR China.
| | - Chaojin Chen
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, PR China.
| | - Shaoli Zhou
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, PR China.
| |
Collapse
|