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
|
Yu Y, Wang T, Li Q, Zhao H, Li B, Lei D, Dong F, Xiao Y, Wang S, Ji Y. DL-3-n-butylphthalide inhibits astrocyte activation in the cortical penumbra of ischemia-reperfusion model rats via AKT signaling. Brain Res Bull 2025; 225:111332. [PMID: 40185418 DOI: 10.1016/j.brainresbull.2025.111332] [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: 01/20/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Ischemic stroke triggers rapid activation of astrocytes, which contributes to tissue damage. Dl-3-n-butylphthalide (NBP), an independently developed compound in China for the treatment of ischemic stroke, has unclear molecular mechanisms. In this study, we established a Sprague-Dawley rat model of middle cerebral artery occlusion (MCAO) by occluding the middle cerebral artery for 1.5 h followed by reperfusion for 72 h. We assessed neurological scores, infarct volume, neuronal injury, and the expression levels of GFAP, C3, S100A10, GLT-1, p-AKT/AKT, and p-mTOR/mTOR, as well as immunofluorescence double staining of C3/S100A10 with GFAP and GLT-1 respectively. NBP significantly improved neurological function in MCAO rats, reduced infarct area, alleviated neuronal injury, inhibited A1 astrocyte polarization, promoted A2 astrocyte polarization, and upregulated GLT-1 expression. However, the AKT inhibitor (TCN) weakened NBP's regulatory effects on astrocytes and GLT-1. Finally, immunofluorescence experiments showed that GLT-1 colocalized more effectively with A2 astrocytes than with A1 astrocytes. We demonstrated that NBP reduces astrocyte activation and upregulates GLT-1 expression via the AKT/mTOR pathway, providing new insights into therapeutic strategies for ischemic stroke and valuable clues for drug design.
Collapse
Affiliation(s)
- Yiwen Yu
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tinghong Wang
- Department of Forensic Pathology, School of Basic Medical Science and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qiuling Li
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hao Zhao
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Biao Li
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Dong Lei
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Fei Dong
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yu Xiao
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shan Wang
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yifei Ji
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China.
| |
Collapse
|
3
|
Jiang H, Zhou F, Guo L, Gao Y, Kong N, Xu M, Zhang F. Implications of hippocampal excitatory amino acid transporter 2 in modulating anxiety and visceral pain in a mouse model of inflammatory bowel disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167832. [PMID: 40203955 DOI: 10.1016/j.bbadis.2025.167832] [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: 12/08/2024] [Revised: 03/22/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic inflammation and significantly impairs quality of life through anxiety-like behaviors and visceral pain. Early evaluation of the risk of anxiety-like behaviors and visceral pain in IBD patients, along with targeted treatment, may benefit disease management. Visceral pain and anxiety-like behavior are often accompanied by neurological damage. Previous studies have shown that abnormal accumulation of glutamate can cause excitatory neurotoxic effects, leading to central nervous system (CNS) damage. Excitatory amino acid transporters (EAATs), particularly EAAT2, are known to regulate glutamate levels. The impact of hippocampal EAAT2 modulation on these clinical features in IBD is yet to be evaluated. Therefore, we designed this experiment to test this hypothesis. This study aimed to investigate the impact of altered levels of hippocampal EAAT2 on anxiety-like behaviors and visceral pain in mice with IBD. We observed reduced EAAT2 expression, increased glutamate levels, elevated N-methyl-d-aspartate receptors (NMDAR) expression, and obvious glutamate toxicity in the hippocampus of dextran sulfate sodium (DSS) induced IBD model mice. These mice exhibited significant visceral pain and anxiety-like behaviors. In summary, the reduced expression of EAAT2 in the hippocampus of individuals with IBD leads to elevated glutamate levels, resulting in neuronal damage and ultimately contributing to visceral pain and anxiety-like behaviors. These findings suggest that EAAT2 could serve as a therapeutic target for neurologically derived IBD symptoms.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310024, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Feini Zhou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Lingnan Guo
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310024, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Yiyuan Gao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Ning Kong
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China.
| | - Fan Zhang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China.
| |
Collapse
|
4
|
Blicharz-Futera K, Kamiński M, Grychowska K, Canale V, Zajdel P. Current development in sulfonamide derivatives to enable CNS-drug discovery. Bioorg Chem 2025; 156:108076. [PMID: 39889550 DOI: 10.1016/j.bioorg.2024.108076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 02/03/2025]
Abstract
The encouraging therapeutic potential of sulfonamide-based derivatives has been unraveled by breakthrough discovery of Paul Ehrlich, who pointed out the possibility of fighting microbes with chemicals. Over the decades, the utility of sulfonamides has expanded beyond antimicrobial agents, revealing their usefulness in many areas of pharmacotherapy, including the treatment of central nervous system (CNS) diseases. Through a detailed analysis of preclinical and clinical data, we identify key sulfonamide-based compounds that have demonstrated significant CNS activity. We also discuss the challenges in the development of sulfonamide derivatives as enzyme/ion channel inhibitors or receptor ligands for CNS applications, describing their mode of action and therapeutic significance. This is followed by the characteristics of pharmacological targets, structure-activity relationships, ADMET properties, efficacy in experimental animal models, and outcomes from clinical trials. Overall, the versatile nature of arylsulfonamides makes them a valuable motif in drug discovery, offering diverse opportunities for the development of novel agents for treating CNS disorders.
Collapse
Affiliation(s)
- Klaudia Blicharz-Futera
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Łazarza Street, 31-530 Krakow, Poland
| | - Michał Kamiński
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Łazarza Street, 31-530 Krakow, Poland
| | - Katarzyna Grychowska
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Vittorio Canale
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Paweł Zajdel
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland.
| |
Collapse
|
5
|
Toti A, Lucarini E, Ferrara V, Parisio C, Ciampi C, Gerace E, Micheli L, Margiotta F, Venturi D, Mello T, Lacal PM, Graziani G, Mannaioni G, Ghelardini C, Di Cesare Mannelli L. The dual role of VEGF-A in a complex in vitro model of oxaliplatin-induced neurotoxicity: Pain-related and neuroprotective effects. Neurotherapeutics 2025; 22:e00532. [PMID: 39939241 PMCID: PMC12014407 DOI: 10.1016/j.neurot.2025.e00532] [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: 08/13/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/14/2025] Open
Abstract
Vascular endothelial growth factor (VEGF)-A is a main player in the development of neuropathic pain induced by chemotherapy and the pharmacological blockade of VEGF receptor (VEGFR) subtype 1 is a pain killer strategy. Interestingly, VEGF-A has been demonstrated to have also neuroprotective properties. The aim of the study was to investigate the neuroprotective role of VEGF-A against oxaliplatin neurotoxicity, attempting to discriminate pain-related and restorative signaling pathways. We used rat organotypic spinal cord slices treated with oxaliplatin, as an in vitro model to study chemotherapy-induced toxicity. In this model, 10 μM oxaliplatin caused a time-dependent release of VEGF-A, which was reduced by the astrocyte inhibitor fluorocitrate. Moreover, glia inhibition exacerbated oxaliplatin-induced cytotoxicity in a VEGF-A sensitive manner. Treatment with VEGF165b, the main isoform of VEGF-A, prevented the oxaliplatin-induced neuronal damage (indicated by NeuN staining) and astrocyte activation (indicated by GFAP staining). In addition, the blockade of VEGFR-2 by the selective antibody DC101 blunted the protective action of VEGF165b. In the same model, VEGF165b increased the release of molecules relevant in pain signaling, like substance P and CGRP, as well as the mRNA expression of glutamate transporters (EAAT1 and EAAT2), similarly to oxaliplatin and these effects were prevented by the selective VEGFR-1 blocker antibody D16F7. In conclusion, VEGF-A plays a dichotomic role in an in vitro model of chemotherapy-induced toxicity, either promoting neuroprotection or triggering pain mediators release, depending on which of its two receptors is activated. The selective management of VEGF-A signaling is suggested as a therapeutic approach.
Collapse
Affiliation(s)
- A Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - E Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - V Ferrara
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - C Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - C Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - E Gerace
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy; Department of Health Sciences, University of Florence, Florence, Italy
| | - L Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy.
| | - F Margiotta
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - D Venturi
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - T Mello
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - G Graziani
- Department of Systems Medicine, Pharmacology Section, University of Rome Tor Vergata, Rome, Italy
| | - G Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - C Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - L Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
6
|
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
|
7
|
Kamiński K, Socała K, Abram M, Jakubiec M, Reeb KL, Temmermand R, Zagaja M, Maj M, Kolasa M, Faron‐Górecka A, Andres‐Mach M, Szewczyk A, Hameed MQ, Fontana ACK, Rotenberg A, Kamiński RM. Enhancement of Glutamate Uptake as Novel Antiseizure Approach: Preclinical Proof of Concept. Ann Neurol 2025; 97:344-357. [PMID: 39512205 PMCID: PMC11740271 DOI: 10.1002/ana.27124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/14/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
OBJECTIVE Excitotoxicity is a common hallmark of epilepsy and other neurological diseases associated with elevated extracellular glutamate levels. Thus, here, we studied the protective effects of (R)-AS-1, a positive allosteric modulator (PAM) of glutamate uptake in epilepsy models. METHODS (R)-AS-1 was evaluated in a range of acute and chronic seizure models, while its adverse effect profile was assessed in a panel of standard tests in rodents. The effect of (R)-AS-1 on glutamate uptake was assessed in COS-7 cells expressing the transporter. WAY 213613, a selective competitive EAAT2 inhibitor, was used to probe the reversal of the enhanced glutamate uptake in the same transporter expression system. Confocal microscopy and Western blotting analyses were used to study a potential influence of (R)-AS-1 on GLT-1 expression in mice. RESULTS (R)-AS-1 showed robust protection in a panel of animal models of seizures and epilepsy, including the maximal electroshock- and 6 Hz-induced seizures, corneal kindling, mesial temporal lobe epilepsy, lamotrigine-resistant amygdala kindling, as well as seizures induced by pilocarpine or Theiler's murine encephalomyelitis virus. Importantly, (R)-AS-1 displayed a favorable adverse effect profile in the rotarod, the minimal motor impairment, and the Irwin tests. (R)-AS-1 enhanced glutamate uptake in vitro and this effect was abolished by WAY 213613, while no influence on GLT-1 expression in vivo was observed after repeated treatment. INTERPRETATION Collectively, our results show that (R)-AS-1 has favorable tolerability and provides robust preclinical efficacy against seizures. Thus, allosteric enhancement of EAAT2 function could offer a novel therapeutic strategy for treatment of epilepsy and potentially other neurological disorders associated with glutamate excitotoxicity. ANN NEUROL 2025;97:344-357.
Collapse
Affiliation(s)
- Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of PharmacyJagiellonian University Medical CollegeKrakowPoland
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and BiotechnologyMaria Curie‐Skłodowska UniversityLublinPoland
| | - Michał Abram
- Department of Medicinal Chemistry, Faculty of PharmacyJagiellonian University Medical CollegeKrakowPoland
| | - Marcin Jakubiec
- Department of Medicinal Chemistry, Faculty of PharmacyJagiellonian University Medical CollegeKrakowPoland
| | - Katelyn L. Reeb
- Department of Pharmacology and PhysiologyDrexel University College of MedicinePhiladelphiaPAUSA
| | - Rhea Temmermand
- Department of Pharmacology and PhysiologyDrexel University College of MedicinePhiladelphiaPAUSA
| | - Mirosław Zagaja
- Department of Experimental PharmacologyInstitute of Rural HealthLublinPoland
| | - Maciej Maj
- Department of BiopharmacyMedical University of LublinLublinPoland
| | - Magdalena Kolasa
- Department of PharmacologyMaj Institute of Pharmacology Polish Academy of SciencesKrakowPoland
| | - Agata Faron‐Górecka
- Department of PharmacologyMaj Institute of Pharmacology Polish Academy of SciencesKrakowPoland
| | - Marta Andres‐Mach
- Department of Experimental PharmacologyInstitute of Rural HealthLublinPoland
| | - Aleksandra Szewczyk
- Department of Experimental PharmacologyInstitute of Rural HealthLublinPoland
| | - Mustafa Q. Hameed
- Department of Neurology, Division of Epilepsy and Clinical Neurophysiology, Neuromodulation ProgramBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- Department of Neurology, F.M. Kirby Neurobiology CenterBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Andréia C. K. Fontana
- Department of Pharmacology and PhysiologyDrexel University College of MedicinePhiladelphiaPAUSA
| | - Alexander Rotenberg
- Department of Neurology, Division of Epilepsy and Clinical Neurophysiology, Neuromodulation ProgramBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- Department of Neurology, F.M. Kirby Neurobiology CenterBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Rafał M. Kamiński
- Department of Medicinal Chemistry, Faculty of PharmacyJagiellonian University Medical CollegeKrakowPoland
| |
Collapse
|
8
|
Shen Y, Zhang X, Liu S, Xin L, Xuan W, Zhuang C, Chen Y, Chen B, Zheng X, Wu R, Lin Y. CEST imaging combined with 1H-MRS reveal the neuroprotective effects of riluzole by improving neurotransmitter imbalances in Alzheimer's disease mice. Alzheimers Res Ther 2025; 17:20. [PMID: 39806490 PMCID: PMC11726951 DOI: 10.1186/s13195-025-01672-3] [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: 10/29/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND The imbalance of glutamate (Glu) and gamma-aminobutyric acid (GABA) neurotransmitter system plays a crucial role in the pathogenesis of Alzheimer's disease (AD). Riluzole is a Glu modulator originally approved for amyotrophic lateral sclerosis that has shown potential neuroprotective effects in various neurodegenerative disorders. However, whether riluzole can improve Glu and GABA homeostasis in AD brain and its related mechanism of action remain unknown. This study utilized chemical exchange saturation transfer (CEST) imaging combined with proton magnetic resonance spectroscopy (1H-MRS) to monitor the dynamic changes of Glu and GABA in riluzole-treated AD mice, aiming to evaluate the efficacy and mechanism of riluzole in AD treatment. METHODS GluCEST, GABACEST and 1H-MRS were used to longitudinally monitor Glu and GABA levels in 3xTg AD mice treated with riluzole (12.5 mg/kg/day) or vehicle for 20 weeks. Magnetic resonance measurements were performed at baseline, 6, 12, and 20 weeks post-treatment. Cognitive performance was assessed using the Morris Water Maze (MWM) at baseline, 10, and 20 weeks. At the study endpoint, immunohistochemistry, Nissl staining, and Western blot were used to evaluate the brain pathology, neuronal survival, and protein expression. RESULTS GluCEST, GABACEST and 1H-MRS consistently revealed higher levels of Glu and GABA in the brain of riluzole-treated AD mice compared to untreated controls, which were associated with improvements in spatial learning and memory. The cognitive improvements significantly correlated with the increased GluCEST signals and Glu levels. Immunohistochemistry and Nissl staining demonstrated that riluzole treatment reduced amyloid-beta (Aβ) deposition, tau hyperphosphorylation, GFAP-positive astrocyte activation, and prevented neuronal loss. Moreover, riluzole upregulated the expression of excitatory amino acid transporter 2 (EAAT2), glutamic acid decarboxylase 65/67 (GAD65/67), and glutamine synthetase (GS), suggesting enhanced neurotransmitter metabolism. CONCLUSIONS CEST imaging combined with 1H-MRS demonstrated the effectiveness of riluzole in modulating Glu- and GABA-related changes and improving cognitive function in 3xTg AD mice, potentially through regulating key proteins involved in neurotransmitter metabolism. These findings suggest riluzole as a therapeutic agent for Alzheimer's disease and highlight the utility of multimodal MR imaging in monitoring treatment response and exploring disease mechanisms.
Collapse
Affiliation(s)
- Yuanyu Shen
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaolei Zhang
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Siqi Liu
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Lijing Xin
- Center for Biomedical Imaging (CIBM), Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Physics (IPHYS), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Wentao Xuan
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Caiyu Zhuang
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yue Chen
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Beibei Chen
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xinhui Zheng
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Renhua Wu
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yan Lin
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
9
|
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
|
10
|
M J VK, Mitteaux J, Wang Z, Wheeler E, Tandon N, Yun Jung S, Hudson RHE, Monchaud D, Tsvetkov AS. Small molecule-based regulation of gene expression in human astrocytes switching on and off the G-quadruplex control systems. J Biol Chem 2025; 301:108040. [PMID: 39615684 PMCID: PMC11750478 DOI: 10.1016/j.jbc.2024.108040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 01/03/2025] Open
Abstract
A great deal of attention is being paid to strategies seeking to uncover the biology of the four-stranded nucleic acid structure G-quadruplex (G4) via their stabilization in cells with G4-specific ligands. The conventional definition of chemical biology implies that a complete assessment of G4 biology can only be achieved by implementing a complementary approach involving the destabilization of cellular G4s by ad hoc molecular effectors. We report here on an unprecedented comparison of the cellular consequences of G4 chemical stabilization by pyridostatin (PDS) and destabilization by phenylpyrrolocytosine (PhpC) at both transcriptome- and proteome-wide scales in patient-derived primary human astrocytes. Our results show that the stabilization of G4s by PDS triggers the dysregulation of many cellular circuitries, the most drastic effects originating in the downregulation of 354 transcripts and 158 proteins primarily involved in RNA transactions. In contrast, destabilization of G4s by PhpC modulates the G4 landscapes in a far more focused manner with upregulation of 295 proteins, mostly involved in RNA transactions as well, thus mirroring the effects of PDS. Our study is the first of its kind to report the extent of G4-associated cellular circuitries in human cells by systematically pitting the effect of G4 stabilization against destabilization in a direct and unbiased manner.
Collapse
Affiliation(s)
- Vijay Kumar M J
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, Texas, USA
| | - Jérémie Mitteaux
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB), CNRS UMR6302, Dijon, France
| | - Zi Wang
- Department of Chemistry, The University of Western Ontario, London, Ontario, Canada
| | - Ellery Wheeler
- The Department of Neurosurgery, The University of Texas, McGovern Medical School at Houston, Houston, Texas, USA
| | - Nitin Tandon
- The Department of Neurosurgery, The University of Texas, McGovern Medical School at Houston, Houston, Texas, USA
| | - Sung Yun Jung
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas, USA
| | - Robert H E Hudson
- Department of Chemistry, The University of Western Ontario, London, Ontario, Canada
| | - David Monchaud
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB), CNRS UMR6302, Dijon, France.
| | - Andrey S Tsvetkov
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, Texas, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; UTHealth Consortium on Aging, The University of Texas McGovern Medical School, Houston, Texas, USA.
| |
Collapse
|
11
|
Hameed MQ, D'Ambrosio R, Eastman C, Hui B, Lin R, Vermudez SAD, Liebhardt A, Choe Y, Klein P, Rundfeldt C, Löscher W, Rotenberg A. A comparison of the antiepileptogenic efficacy of two rationally chosen multitargeted drug combinations in a rat model of posttraumatic epilepsy. Exp Neurol 2024; 382:114962. [PMID: 39288831 DOI: 10.1016/j.expneurol.2024.114962] [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: 06/19/2024] [Revised: 08/08/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Post-traumatic epilepsy (PTE) is a recurrent and often drug-refractory seizure disorder caused by traumatic brain injury (TBI). No single drug treatment prevents PTE, but preventive drug combinations that may prophylax against PTE have not been studied. Based on a systematic evaluation of rationally chosen drug combinations in the intrahippocampal kainate (IHK) mouse model of acquired epilepsy, we identified two multi-targeted drug cocktails that exert strong antiepileptogenic effects. The first, a combination of levetiracetam (LEV) and topiramate, only partially prevented spontaneous recurrent seizures in the model. We therefore added atorvastatin (ATV) to the therapeutic cocktail (TC) to increase efficacy, forming "TC-001". The second cocktail - a combination of LEV, ATV, and ceftriaxone, termed "TC-002" - completely prevented epilepsy in the mouse IHK model. In the present proof-of-concept study, we tested whether the two drug cocktails prevent epilepsy in a rat PTE model in which recurrent electrographic seizures develop after severe rostral parasagittal fluid percussion injury (FPI). Following FPI, rats were either treated over 3-4 weeks with vehicle or drug cocktails, starting either 1 or 4-6 h after the injury. Using mouse doses of TC-001 and TC-002, no significant antiepileptogenic effect was obtained in the rat PTE model. However, when using allometric scaling of drug doses to consider the differences in body surface area between mice and rats, PTE was prevented by TC-002. Furthermore, the latter drug cocktail partially prevented the loss of perilesional cortical parvalbumin-positive GABAergic interneurons. Plasma and brain drug analysis showed that these effects of TC-002 occurred at clinically relevant levels of the individual TC-002 drug components. In silico analysis of drug-drug brain protein interactions by the STITCH database indicated that TC-002 impacts a larger functional network of epilepsy-relevant brain proteins than each drug alone, providing a potential network pharmacology explanation for the observed antiepileptogenic and neuroprotective effects observed with this combination.
Collapse
Affiliation(s)
- Mustafa Q Hameed
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raimondo D'Ambrosio
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Cliff Eastman
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Benjamin Hui
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rui Lin
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sheryl Anne D Vermudez
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amanda Liebhardt
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yongho Choe
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pavel Klein
- PrevEp, Inc., Bethesda, MD, USA; Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | | | - Wolfgang Löscher
- PrevEp, Inc., Bethesda, MD, USA; Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany.
| | - Alexander Rotenberg
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; PrevEp, Inc., Bethesda, MD, USA.
| |
Collapse
|
12
|
Xie D, Zhang P, You S, Shen Y, Xu W, Zhan C, Zhang J. Salidroside derivative SHPL-49 attenuates glutamate excitotoxicity in acute ischemic stroke via promoting NR2A-CAMKⅡα-Akt /CREB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155583. [PMID: 39173548 DOI: 10.1016/j.phymed.2024.155583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Ischemic stroke is a significant cause of death and disability with a limited treatment time window. The reduction of early glutamate excitotoxicity using neuroprotective agents targeting N-methyl-d-aspartic acid (NMDA) receptors have attracted recent research attention. SHPL-49, a structurally modified derivative of salidroside, was synthesized by our team. Previous studies have confirmed the neuroprotective efficacy of SHPL-49 in rats with ischemic stroke. However, the underlying mechanisms need to be clarified. METHODS We conducted in vivo experiments using the permanent middle cerebral artery occlusion rat model to investigate the role of SHPL-49 in glutamate release at different time points and treatment durations. Glutamate transporters and receptor proteins and neural survival proteins in the brain were also examined at the same time points. In vitro, primary neurons and the coculture system of primary neurons-astrocytes were subjected to oxygen-glucose deprivation and glutamate injury. Proteomics and parallel reaction monitoring analyses were performed to identify potential therapeutic targets of SHPL-49, which were further confirmed through in vitro experiments on the inhibition and mutation of the target. RESULTS SHPL-49 significantly reduced glutamate release caused by hypoxia-ischemia. One therapeutic pathway of SHPL-49 was promoting the expression of glutamate transporter-1 to increase glutamate reuptake and further reduce the occurrence of subsequent neurotoxicity. In addition, we explored the therapeutic targets of SHPL-49 and its regulatory effects on glutamate receptors for the first time. SHPL-49 enhanced neuroprotection by activating the NMDA subunit NR2A, which upregulated the cyclic-AMP response binding protein (CREB) neural survival pathway and Akt phosphorylation. Since calcium/calmodulin-dependent kinase IIα (CaMKIIα) is necessary for synaptic transmission of NMDA receptors, we explored the interaction between CaMKIIα and SHPL-49, which protected CaMKIIα from hypoxia-ischemia-induced autophosphorylation damage. CONCLUSION Overall, SHPL-49 enhanced neuronal survival and attenuated acute ischemic stroke by promoting the NR2A-CAMKⅡα-Akt/CREB pathway. Our study provides the first evidence demonstrating that the neuroprotective effect of SHPL-49 is achieved by promoting the NR2A subunit to extend the treatment time window, making it a promising drug for ischemic stroke.
Collapse
Affiliation(s)
- Dong Xie
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, No.1200 Cailun Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Pei Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, No.1200 Cailun Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Suxin You
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, No.1200 Cailun Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Yue Shen
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, No.1200 Cailun Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Wenwen Xu
- Shanghai Hutchison Pharmaceuticals Co., Ltd, Shanghai 201400, China
| | - Changsen Zhan
- Shanghai Hutchison Pharmaceuticals Co., Ltd, Shanghai 201400, China
| | - Jiange Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, No.1200 Cailun Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China.
| |
Collapse
|
13
|
Zhang K, Yang Z, Yang Z, Du L, Zhou Y, Fu S, Wang X, Li X, Liu D, He X. The m6A reader YTHDC2 promotes the pathophysiology of temporal lobe epilepsy by modulating SLC7A11-dependent glutamate dysregulation in astrocytes. Theranostics 2024; 14:5551-5570. [PMID: 39310099 PMCID: PMC11413790 DOI: 10.7150/thno.100703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
Rationale: Epilepsy affects over 70 million people globally, with temporal lobe epilepsy with hippocampal sclerosis (TLE-HS) often progressing to a drug-resistant state. Recent research has highlighted the role of reactive astrocytes and glutamate dysregulation in epilepsy pathophysiology. This study aims to investigate the involvement of astrocytic xCT, a glutamate-cystine antiporter, and its regulation by the m6A reader protein YTHDC2 in TLE-HS. Methods: A pilocarpine-induced epilepsy model in mice was used to study the role of xCT in reactive astrocytes. The expression of xCT and its regulation by YTHDC2 were assessed through various molecular and cellular techniques. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting were used to measure mRNA and protein levels of xCT and YTHDC2, respectively; immunofluorescence was utilized to visualize their localization and expression in astrocytes. In vivo glutamate measurements were conducted using microdialysis to monitor extracellular glutamate levels in the hippocampus. RNA immunoprecipitation-qPCR (RIP-qPCR) was performed to investigate the binding of YTHDC2 to SLC7A11 mRNA, while methylated RNA immunoprecipitation-qPCR (MeRIP-qPCR) was performed to quantify m6A modifications on SLC7A11 mRNA. A dual-luciferase reporter assay was conducted to assess the effect of m6A modifications on SLC7A11 mRNA translation, and polysome profiling was employed to evaluate the translational efficiency of SLC7A11 mRNA. Inhibition experiments involved shRNA-mediated knockdown of SLC7A11 (commonly known as xCT) and YTHDC2 expression in astrocytes. Video-electroencephalogram (EEG) recordings were used to monitor seizure activity in mice. Results: The xCT transporter in reactive astrocytes significantly contributes to elevated extracellular glutamate levels, enhancing neuronal excitability and seizure activity. Increased xCT expression is influenced by the m6A reader protein YTHDC2, which regulates its expression through m6A methylation. Inhibition of xCT or YTHDC2 in astrocytes reduces glutamate levels and effectively controls seizures in a mouse model. Specifically, mice with SLC7A11- or YTHDC2-knockdown astrocytes showed decreased glutamate concentration in the hippocampus and reduced frequency and duration of epileptic seizures. Conclusions: This study highlights the therapeutic potential of targeting YTHDC2 and xCT in reactive astrocytes to mitigate epilepsy. The findings provide a novel perspective on the mechanisms of glutamate dysregulation and their implications in seizure pathophysiology, suggesting that modulation of YTHDC2 and xCT could be a promising strategy for treating TLE.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiquan Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuanyi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liangchao Du
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shiyu Fu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyue Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xing Li
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha 410078, Hunan Province, China
| | - Dingyang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinghui He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Galli A, Moretti S, Dule N, Di Cairano ES, Castagna M, Marciani P, Battaglia C, Bertuzzi F, Fiorina P, Pastore I, La Rosa S, Davalli A, Folli F, Perego C. Hyperglycemia impairs EAAT2 glutamate transporter trafficking and glutamate clearance in islets of Langerhans: implications for type 2 diabetes pathogenesis and treatment. Am J Physiol Endocrinol Metab 2024; 327:E27-E41. [PMID: 38690938 PMCID: PMC11390119 DOI: 10.1152/ajpendo.00069.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Abstract
Pancreatic endocrine cells employ a sophisticated system of paracrine and autocrine signals to synchronize their activities, including glutamate, which controls hormone release and β-cell viability by acting on glutamate receptors expressed by endocrine cells. We here investigate whether alteration of the excitatory amino acid transporter 2 (EAAT2), the major glutamate clearance system in the islet, may occur in type 2 diabetes mellitus and contribute to β-cell dysfunction. Increased EAAT2 intracellular localization was evident in islets of Langerhans from T2DM subjects as compared with healthy control subjects, despite similar expression levels. Chronic treatment of islets from healthy donors with high-glucose concentrations led to the transporter internalization in vesicular compartments and reduced [H3]-d-glutamate uptake (65 ± 5% inhibition), phenocopying the findings in T2DM pancreatic sections. The transporter relocalization was associated with decreased Akt phosphorylation protein levels, suggesting an involvement of the phosphoinositide 3-kinase (PI3K)/Akt pathway in the process. In line with this, PI3K inhibition by a 100-µM LY294002 treatment in human and clonal β-cells caused the transporter relocalization in intracellular compartments and significantly reduced the glutamate uptake compared to control conditions, suggesting that hyperglycemia changes the trafficking of the transporter to the plasma membrane. Upregulation of the glutamate transporter upon treatment with the antibiotic ceftriaxone rescued hyperglycemia-induced β-cells dysfunction and death. Our data underscore the significance of EAAT2 in regulating islet physiology and provide a rationale for potential therapeutic targeting of this transporter to preserve β-cell survival and function in diabetes.NEW & NOTEWORTHY The glutamate transporter SLC1A2/excitatory amino acid transporter 2 (EAAT2) is expressed on the plasma membrane of pancreatic β-cells and controls islet glutamate clearance and β-cells survival. We found that the EAAT2 membrane expression is lost in the islets of Langerhans from type 2 diabetes mellitus (T2DM) patients due to hyperglycemia-induced downregulation of the phosphoinositide 3-kinase/Akt pathway and modification of its intracellular trafficking. Pharmacological rescue of EAAT2 expression prevents β-cell dysfunction and death, suggesting EAAT2 as a new potential target of intervention in T2DM.
Collapse
Affiliation(s)
- Alessandra Galli
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Stefania Moretti
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nevia Dule
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Eliana Sara Di Cairano
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Michela Castagna
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paola Marciani
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Cristina Battaglia
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | | | - Paolo Fiorina
- Department of Biomedical and Clinical Sciences "L. Sacco,"Università degli Studi di Milano, Milan, Italy
- Endocrinology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Ida Pastore
- Endocrinology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Stefano La Rosa
- Unit of Pathology, Department of Oncology, ASST Sette Laghi, Varese, Italy
- Department of Medicine and Technological Innovation, Università degli Studi dell'Insubria, Varese, Italy
| | - Alberto Davalli
- Diabetes and Endocrinology Unit, Department of Internal Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Franco Folli
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Carla Perego
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
15
|
陈 洁, 刘 晨, 王 春, 李 丽, 陶 伟, 徐 婧, 唐 红, 黄 丽. [Exogenous leptin improves cerebral ischemia-reperfusion-induced glutamate excitotoxic injury in mice by up-regulating GLT-1 and GLAST expression in astrocytes]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1079-1087. [PMID: 38977337 PMCID: PMC11237293 DOI: 10.12122/j.issn.1673-4254.2024.06.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Indexed: 07/10/2024]
Abstract
OBJECTIVE To investigate the protective effect of exogenous leptin against focal cerebral ischemia-reperfusion (I/R) injury in mice and explore the underlying mechanism. METHODS A total of 100 C57BL/6 mice were randomly divided into 5 groups, including a sham-operated group, cerebral I/R model group, and 3 leptin treatment groups with intraperitoneal injections of 0.5, 1.0 or 2.0 leptin immediately after occlusion of the internal carotid artery. At 24 h after reperfusion, neurological function scores of the mice were assessed, and TTC staining was used to determine the area of cerebral infarction. The pathological changes in the cortical brain tissue of the mice were observed using HE staining, and degenerative damage of the cortical neurons were assessed with Fluoro-Jade C staining. The expression of glial fibrillary acidic protein in cortical brain tissues was detected using immunohistochemistry and Western blotting. In another 45 C57BL/6 mice with sham operation, I/R modeling, or leptin (1 mg/kg) treatment, glutamic acid in the cortical brain tissue was detected using glutamate assay, and cortical glutamate-aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1) protein expressions were detected using immunohistochemistry. RESULTS Compared with the I/R model mice, the leptin-treated mice had significantly lower neurological deficit scores, smaller cerebral infarct area, milder pathologies in the cortical brain tissue, and lessened cortical neuronal damage with normal morphology and less excessive proliferation of the astrocytes. Leptin treatment significantly up-regulated the expressions of GLT-1 and GLAST and lowered the content of glutamic acid in the brain tissue of the I/R mice. CONCLUSION Exogenous leptin has obvious neuroprotective effect against cerebral I/R injury in mice, mediated probably by controlling excessive astrocyte proliferation and up-regulating cortical GLT-1 and GLAST expressions to reduce glutamate-mediated excitotoxic injury of the astrocytes.
Collapse
|
16
|
Ohki CMY, Benazzato C, van der Linden V, França JV, Toledo CM, Machado RRG, Araujo DB, Oliveira DBL, Neris RS, Assunção-Miranda I, de Oliveira Souza IN, Nogueira CO, Leite PEC, van der Linden H, Figueiredo CP, Durigon EL, Clarke JR, Russo FB, Beltrão-Braga PCB. Zika virus infection impairs synaptogenesis, induces neuroinflammation, and could be an environmental risk factor for autism spectrum disorder outcome. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167097. [PMID: 38408544 DOI: 10.1016/j.bbadis.2024.167097] [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: 08/14/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Zika virus (ZIKV) infection was first associated with Central Nervous System (CNS) infections in Brazil in 2015, correlated with an increased number of newborns with microcephaly, which ended up characterizing the Congenital Zika Syndrome (CZS). Here, we investigated the impact of ZIKV infection on the functionality of iPSC-derived astrocytes. Besides, we extrapolated our findings to a Brazilian cohort of 136 CZS children and validated our results using a mouse model. Interestingly, ZIKV infection in neuroprogenitor cells compromises cell migration and causes apoptosis but does not interfere in astrocyte generation. Moreover, infected astrocytes lost their ability to uptake glutamate while expressing more glutamate transporters and secreted higher levels of IL-6. Besides, infected astrocytes secreted factors that impaired neuronal synaptogenesis. Since these biological endophenotypes were already related to Autism Spectrum Disorder (ASD), we extrapolated these results to a cohort of children, now 6-7 years old, and found seven children with ASD diagnosis (5.14 %). Additionally, mice infected by ZIKV revealed autistic-like behaviors, with a significant increase of IL-6 mRNA levels in the brain. Considering these evidence, we inferred that ZIKV infection during pregnancy might lead to synaptogenesis impairment and neuroinflammation, which could increase the risk for ASD.
Collapse
Affiliation(s)
| | - Cecília Benazzato
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Julia V França
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carmen M Toledo
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Romulo S Neris
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iranaia Assunção-Miranda
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Clara O Nogueira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo Emilio Corrêa Leite
- Clinical Research Unit of the Antonio Pedro Hospital, Federal Fluminense University, Rio de Janeiro, Brazil
| | | | - Claudia P Figueiredo
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edison Luiz Durigon
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Institut Pasteur de São Paulo, São Paulo, Brazil
| | - Julia R Clarke
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
17
|
Shukla H, John D, Banerjee S, Tiwari AK. Drug repurposing for neurodegenerative diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:249-319. [PMID: 38942541 DOI: 10.1016/bs.pmbts.2024.03.035] [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: 06/30/2024]
Abstract
Neurodegenerative diseases (NDDs) are neuronal problems that include the brain and spinal cord and result in loss of sensory and motor dysfunction. Common NDDs include Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Multiple Sclerosis (MS), and Amyotrophic Lateral Sclerosis (ALS) etc. The occurrence of these diseases increases with age and is one of the challenging problems among elderly people. Though, several scientific research has demonstrated the key pathologies associated with NDDs still the underlying mechanisms and molecular details are not well understood and need to be explored and this poses a lack of effective treatments for NDDs. Several lines of evidence have shown that NDDs have a high prevalence and affect more than a billion individuals globally but still, researchers need to work forward in identifying the best therapeutic target for NDDs. Thus, several researchers are working in the directions to find potential therapeutic targets to alter the disease pathology and treat the diseases. Several steps have been taken to identify the early detection of the disease and drug repurposing for effective treatment of NDDs. Moreover, it is logical that current medications are being evaluated for their efficacy in treating such disorders; therefore, drug repurposing would be an efficient, safe, and cost-effective way in finding out better medication. In the current manuscript we discussed the utilization of drugs that have been repurposed for the treatment of AD, PD, HD, MS, and ALS.
Collapse
Affiliation(s)
- Halak Shukla
- Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India
| | - Diana John
- Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India
| | - Shuvomoy Banerjee
- Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India
| | - Anand Krishna Tiwari
- Genetics and Developmental Biology Laboratory, Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India.
| |
Collapse
|
18
|
Jiang J, Wang L, Li Q, Wang Y, Wang Z. HIV-1 gp120 amplifies astrocyte elevated gene-1 activity to compromise the integrity of the outer blood-retinal barrier. AIDS 2024; 38:779-789. [PMID: 38578957 DOI: 10.1097/qad.0000000000003844] [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: 04/07/2024]
Abstract
OBJECTIVE This study aims to investigate the functions and mechanistic pathways of Astrocyte Elevated Gene-1 (AEG-1) in the disruption of the blood-retinal barrier (BRB) caused by the HIV-1 envelope glycoprotein gp120. DESIGN We utilized ARPE-19 cells challenged with gp120 as our model system. METHODS Several analytical techniques were employed to decipher the intricate interactions at play. These included PCR, Western blot, and immunofluorescence assays for the molecular characterization, and transendothelial electrical resistance (TEER) measurements to evaluate barrier integrity. RESULTS We observed that AEG-1 expression was elevated, whereas the expression levels of tight junction proteins ZO-1, Occludin, and Claudin5 were downregulated in gp120-challenged cells. TEER measurements corroborated these findings, indicating barrier dysfunction. Additional mechanistic studies revealed that the activation of NFκB and MMP2/9 pathways mediated the AEG-1-induced barrier destabilization. Through the use of lentiviral vectors, we engineered cell lines with modulated AEG-1 expression levels. Silencing AEG-1 alleviated gp120-induced downregulation of tight junction proteins and barrier impairment while concurrently inhibiting the NFκB and MMP2/9 pathways. Conversely, overexpression of AEG-1 exacerbated these pathological changes, further compromising the integrity of the BRB. CONCLUSION Gp120 upregulates the expression of AEG-1 and activates the NFκB and MMP2/9 pathways. This in turn leads to the downregulation of tight junction proteins, resulting in the disruption of barrier function.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Ophthalmology, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
19
|
Fontana ACK, Poli AN, Gour J, Srikanth YV, Anastasi N, Ashok D, Khatiwada A, Reeb KL, Cheng MH, Bahar I, Rawls SM, Salvino JM. Synthesis and Structure-Activity Relationships for Glutamate Transporter Allosteric Modulators. J Med Chem 2024; 67:6119-6143. [PMID: 38626917 PMCID: PMC11056993 DOI: 10.1021/acs.jmedchem.3c01909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/23/2024] [Accepted: 03/27/2024] [Indexed: 04/19/2024]
Abstract
Excitatory amino acid transporters (EAATs) are essential CNS proteins that regulate glutamate levels. Excess glutamate release and alteration in EAAT expression are associated with several CNS disorders. Previously, we identified positive allosteric modulators (PAM) of EAAT2, the main CNS transporter, and have demonstrated their neuroprotective properties in vitro. Herein, we report on the structure-activity relationships (SAR) for the analogs identified from virtual screening and from our medicinal chemistry campaign. This work identified several selective EAAT2 positive allosteric modulators (PAMs) such as compounds 4 (DA-023) and 40 (NA-014) from a library of analogs inspired by GT949, an early generation compound. This series also provides nonselective EAAT PAMs, EAAT inhibitors, and inactive compounds that may be useful for elucidating the mechanism of EAAT allosteric modulation.
Collapse
Affiliation(s)
- Andréia C. K. Fontana
- Department
of Pharmacology and Physiology, Drexel University
College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Adi N.R. Poli
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Jitendra Gour
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Yellamelli V.V. Srikanth
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Nicholas Anastasi
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Devipriya Ashok
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Apeksha Khatiwada
- Department
of Pharmacology and Physiology, Drexel University
College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Katelyn L. Reeb
- Department
of Pharmacology and Physiology, Drexel University
College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Mary Hongying Cheng
- Laufer
Center for Physical & Quantitative Biology, Stony Brook University, Stony
Brook, New York 11794, United States
| | - Ivet Bahar
- Department
of Biochemistry and Cell Biology, College of Arts & Sciences and
School of Medicine, Stony Brook University, Stony Brook, New York 11794, United States
- Laufer
Center for Physical & Quantitative Biology, Stony Brook University, Stony
Brook, New York 11794, United States
| | - Scott M. Rawls
- Center
for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140United States
| | - Joseph M. Salvino
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
- The
Wistar
Cancer Center Molecular Screening, The Wistar
Institute, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
20
|
Zhang LY, Hu YY, Liu XY, Wang XY, Li SC, Zhang JG, Xian XH, Li WB, Zhang M. The Role of Astrocytic Mitochondria in the Pathogenesis of Brain Ischemia. Mol Neurobiol 2024; 61:2270-2282. [PMID: 37870679 DOI: 10.1007/s12035-023-03714-z] [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: 06/23/2023] [Accepted: 10/03/2023] [Indexed: 10/24/2023]
Abstract
The morbidity rate of ischemic stroke is increasing annually with the growing aging population in China. Astrocytes are ubiquitous glial cells in the brain and play a crucial role in supporting neuronal function and metabolism. Increasing evidence shows that the impairment or loss of astrocytes contributes to neuronal dysfunction during cerebral ischemic injury. The mitochondrion is increasingly recognized as a key player in regulating astrocyte function. Changes in astrocytic mitochondrial function appear to be closely linked to the homeostasis imbalance defects in glutamate metabolism, Ca2+ regulation, fatty acid metabolism, reactive oxygen species, inflammation, and copper regulation. Here, we discuss the role of astrocytic mitochondria in the pathogenesis of brain ischemic injury and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Xi-Yun Liu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Xiao-Yu Wang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Shi-Chao Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, People's Republic of China.
| |
Collapse
|
21
|
Lee SG. Editorial: 15 Years of Frontiers in Cellular Neuroscience: astrocytes in brain disease. Front Cell Neurosci 2024; 18:1374172. [PMID: 38370033 PMCID: PMC10870418 DOI: 10.3389/fncel.2024.1374172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/20/2024] Open
Affiliation(s)
- Seok-Geun Lee
- Department of Biomedical Science and Technology, BioNanocomposite Research Center, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Zhang D, Hua Z, Li Z. The role of glutamate and glutamine metabolism and related transporters in nerve cells. CNS Neurosci Ther 2024; 30:e14617. [PMID: 38358002 PMCID: PMC10867874 DOI: 10.1111/cns.14617] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/15/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Glutamate and glutamine are the most abundant amino acids in the blood and play a crucial role in cell survival in the nervous system. Various transporters found in cell and mitochondrial membranes, such as the solute carriers (SLCs) superfamily, are responsible for maintaining the balance of glutamate and glutamine in the synaptic cleft and within cells. This balance affects the metabolism of glutamate and glutamine as non-essential amino acids. AIMS This review aims to provide an overview of the transporters and enzymes associated with glutamate and glutamine in neuronal cells. DISCUSSION We delve into the function of glutamate and glutamine in the nervous system by discussing the transporters involved in the glutamate-glutamine cycle and the key enzymes responsible for their mutual conversion. Additionally, we highlight the role of glutamate and glutamine as carbon and nitrogen donors, as well as their significance as precursors for the synthesis of reduced glutathione (GSH). CONCLUSION Glutamate and glutamine play a crucial role in the brain due to their special effects. It is essential to focus on understanding glutamate and glutamine metabolism to comprehend the physiological behavior of nerve cells and to treat nervous system disorders and cancer.
Collapse
Affiliation(s)
- Dongyang Zhang
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Zhongyan Hua
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Zhijie Li
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
23
|
Vyas A, Doshi G. A cross talk on the role of contemporary biomarkers in depression. Biomarkers 2024; 29:18-29. [PMID: 38261718 DOI: 10.1080/1354750x.2024.2308834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/14/2024] [Indexed: 01/25/2024]
Abstract
Introduction: Biomarkers can be used to identify determinants of response to various treatments of mental disorders. Evidence to date demonstrates that markers of inflammatory, neurotransmitter, neurotrophic, neuroendocrine, and metabolic function can predict the psychological and physical consequences of depression in individuals, allowing for the development of new therapeutic targets with fewer side effects. Extensive research has included hundreds of potential biomarkers of depression, but their roles in depression, abnormal patients, and how bioinformatics can be used to improve diagnosis, treatment, and prognosis have not been determined or defined. To determine which biomarkers can and cannot be used to predict treatment response, classify patients for specific treatments, and develop targets for new interventions, proprietary strategies, and current research projects need to be tailored.Material and Methods: This review article focuses on - biomarker systems that would help in the further development and expansion of newer targets - which holds great promise for reducing the burden of depression.Results and Discussion: Further, this review point to the inflammatory response, metabolic marker, and microribonucleic acids, long non-coding RNAs, HPA axis which are - related to depression and can serve as future targets.
Collapse
Affiliation(s)
- Aditi Vyas
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
24
|
Jensen BK. Astrocyte-Neuron Interactions Contributing to Amyotrophic Lateral Sclerosis Progression. ADVANCES IN NEUROBIOLOGY 2024; 39:285-318. [PMID: 39190080 DOI: 10.1007/978-3-031-64839-7_12] [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: 08/28/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex disease impacting motor neurons of the brain, brainstem, and spinal cord. Disease etiology is quite heterogeneous with over 40 genes causing the disease and a vast ~90% of patients having no prior family history. Astrocytes are major contributors to ALS, particularly through involvement in accelerating disease progression. Through study of genetic forms of disease including SOD1, TDP43, FUS, C9orf72, VCP, TBK1, and more recently patient-derived cells from sporadic individuals, many biological mechanisms have been identified to cause intrinsic or glial-mediated neurotoxicity to motor neurons. Overall, many of the normally supportive and beneficial roles that astrocytes contribute to neuronal health and survival instead switch to become deleterious and neurotoxic. While the exact pathways may differ based on disease-origin, altered astrocyte-neuron communication is a common feature of ALS. Within this chapter, distinct genetic forms are examined in detail, along with what is known from sporadic patient-derived cells. Overall, this chapter highlights the interplay between astrocytes and neurons in this complex disease and describes the key features underlying: astrocyte-mediated motor neuron toxicity, excitotoxicity, oxidative/nitrosative stress, protein dyshomeostasis, metabolic imbalance, inflammation, trophic factor withdrawal, blood-brain/blood-spinal cord barrier involvement, disease spreading, and the extracellular matrix/cell adhesion/TGF-β signaling pathways.
Collapse
Affiliation(s)
- Brigid K Jensen
- Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Buccoliero AM, Caporalini C, Moscardi S, Cetica V, Mei D, Conti V, Nozzoli F, Bonaudo C, Battista F, Giordano F, Mura R, Spacca B, Mussa F, D'Onofrio V, Guerrini R, Genitori L, Scagnet M. Leat-associated seizures the possible role of EAAT2, pyruvate carboxylase and glutamine synthetase. Epilepsy Res 2024; 199:107258. [PMID: 38086219 DOI: 10.1016/j.eplepsyres.2023.107258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Drug-resistant epilepsy is a common condition in patients with brain neoplasms. The pathogenesis of tumor-associated seizures is poorly understood. Among the possible pathogenetic mechanisms, the increase in glutamate concentration has been proposed. Glutamate transporters, glutamine synthetase and pyruvate carboxylase are involved in maintaining the physiological concentration of glutamate in the intersynaptic spaces. In our previous research on angiocentric gliomas, we demonstrated that all tumors lacked the expression of the main glutamate transporter EAAT2, while the expression of glutamine synthetase and pyruvate carboxylase was mostly preserved. METHODS In the present study, we evaluated the immunohistochemical expression of EAAT2, glutamine synthetase and pyruvate carboxylase in a heterogeneous series of 25 long-term epilepsy-associated tumors (10 dysembryoplastic neuroepithelial tumors, 7 gangliogliomas, 3 subependymal giant cell astrocytomas, 3 rosette forming glioneuronal tumors, 1 diffuse astrocytoma MYB- or MYBL1-altered and 1 angiocentric glioma). In order to evaluate the incidence of variants in the SLC1A2 gene, encoding EAAT2, in a large number of central nervous system tumors we also queried the PedcBioPortal. RESULTS EAAT2 protein expression was lost in 9 tumors (36 %: 3 dysembryoplastic neuroepithelial tumors, 1 ganglioglioma, 3 subependymal giant cell astrocytomas, 1 diffuse astrocytoma MYB- or MYBL1-altered and 1 angiocentric glioma). Glutamine synthetase protein expression was completely lost in 2 tumors (8 %; 1 ganglioglioma and 1 diffuse astrocytoma MYB- or MYBL1-altered). All tumors of our series but rosette forming glioneuronal tumors (in which neurocytic cells were negative) were diffusely positive for pyruvate carboxylase. Consultation of the PedcBioPortal revealed that of 2307 pediatric brain tumors of different histotype and grade, 20 (< 1%) had variants in the SLC1A2 gene. Among the SLC1A2-mutated tumors, there were no angiocentric gliomas or other LEATs CONCLUSIONS: In conclusion, unlike angiocentric gliomas where the EAAT2 loss is typical and constant, the current study shows the loss of EAAT2 expression only in a fraction of the LEATs. In these cases, we may hypothesize some possible epileptogenic role of the EAAT2 loss. The retained expression of pyruvate carboxylase may contribute to determining a pathological glutamate excess unopposed by glutamine synthetase that resulted expressed to a variable extent in the majority of the tumors. Furthermore, we can assume that the EAAT2 loss in brain tumors in general and in LEATs in particular is more conceivably epigenetic.
Collapse
Affiliation(s)
| | | | - Selene Moscardi
- Pathology Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Valentina Cetica
- Department of Neurosciences, Psychology, Drug Research, and Child Health (NEUROFARBA), University of Florence, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Davide Mei
- Department of Neurosciences, Psychology, Drug Research, and Child Health (NEUROFARBA), University of Florence, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Valerio Conti
- Department of Neurosciences, Psychology, Drug Research, and Child Health (NEUROFARBA), University of Florence, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Filippo Nozzoli
- Histopathology and Molecular Diagnostics, Careggi University Hospital, Florence, Italy
| | - Camilla Bonaudo
- Department of Neurosciences, Psychology, Drug Research, and Child Health (NEUROFARBA), Careggi University Hospital, Florence, Italy
| | - Francesca Battista
- Department of Neurosciences, Psychology, Drug Research, and Child Health (NEUROFARBA), Careggi University Hospital, Florence, Italy
| | - Flavio Giordano
- Department of Neurosciences, Psychology, Drug Research, and Child Health (NEUROFARBA), University of Florence, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Regina Mura
- Department of Neurosurgery, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Barbara Spacca
- Department of Neurosurgery, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Federico Mussa
- Department of Neurosurgery, Meyer Children's Hospital IRCCS, Florence, Italy
| | | | - Renzo Guerrini
- Department of Neurosciences, Psychology, Drug Research, and Child Health (NEUROFARBA), University of Florence, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Lorenzo Genitori
- Department of Neurosciences, Psychology, Drug Research, and Child Health (NEUROFARBA), Careggi University Hospital, Florence, Italy
| | - Mirko Scagnet
- Department of Neurosurgery, Meyer Children's Hospital IRCCS, Florence, Italy
| |
Collapse
|
26
|
Khananshvili D. Neuronal and astrocyte NCX isoform/splice variants: How do they participate in Na + and Ca 2+ signalling? Cell Calcium 2023; 116:102818. [PMID: 37918135 DOI: 10.1016/j.ceca.2023.102818] [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: 10/16/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/04/2023]
Abstract
NCX1, NCX2, and NCX3 gene isoforms and their splice variants are characteristically expressed in different regions of the brain. The tissue-specific splice variants of NCX1-3 isoforms show specific expression profiles in neurons and astrocytes, whereas the relevant NCX isoform/splice variants exhibit diverse allosteric modes of Na+- and Ca2+-dependent regulation. In general, overexpression of NCX1-3 genes leads to neuroprotective effects, whereas their ablation gains the opposite results. At this end, the partial contributions of NCX isoform/splice variants to neuroprotective effects remain unresolved. The glutamate-dependent Na+ entry generates Na+ transients (in response to neuronal cell activities), whereas the Na+-driven Ca2+ entry (through the reverse NCX mode) raises Ca2+ transients. This special mode of signal coupling translates Na+ transients into the Ca2+ signals while being a part of synaptic neurotransmission. This mechanism is of general interest since disease-related conditions (ischemia, metabolic stress, and stroke among many others) trigger Na+ and Ca2+ overload with deadly outcomes of downstream apoptosis and excitotoxicity. The recently discovered mechanisms of NCX allosteric regulation indicate that some NCX variants might play a critical role in the dynamic coupling of Na+-driven Ca2+ entry. In contrast, the others are less important or even could be dangerous under altered conditions (e.g., metabolic stress). This working hypothesis can be tested by applying advanced experimental approaches and highly focused computational simulations. This may allow the development of structure-based blockers/activators that can selectively modulate predefined NCX variants to lessen the life-threatening outcomes of excitotoxicity, ischemia, apoptosis, metabolic deprivation, brain injury, and stroke.
Collapse
Affiliation(s)
- Daniel Khananshvili
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
27
|
Palpagama T, Mills AR, Ferguson MW, Vikas Ankeal P, Turner C, Tippett L, van der Werf B, Waldvogel HJ, Faull RLM, Kwakowsky A. Microglial and Astrocytic Responses in the Human Midcingulate Cortex in Huntington's Disease. Ann Neurol 2023; 94:895-910. [PMID: 37528539 DOI: 10.1002/ana.26753] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVE Patients with Huntington's disease can present with variable difficulties of motor functioning, mood, and cognition. Neurodegeneration occurs in the anterior cingulate cortex of some patients with Huntington's disease and is linked to the presentation of mood symptomatology. Neuroinflammation, perpetrated by activated microglia and astrocytes, has been reported in Huntington's disease and may contribute to disease progression and presentation. This study sought to quantify the density of mutant huntingtin protein and neuroinflammatory glial changes in the midcingulate cortex of postmortem patients with Huntington's disease and determine if either correlates with the presentation of mood, motor, or mixed symptomatology. METHODS Free-floating immunohistochemistry quantified 1C2 immunolabeling density as an indicative marker of mutant huntingtin protein, and protein and morphological markers of astrocyte (EAAT2, Cx43, and GFAP), and microglial (Iba1 and HLA-DP/DQ/DR) activation. Relationships among the level of microglial activation, mutant huntingtin burden, and case characteristics were explored using correlative analysis. RESULTS We report alterations in activated microglia number and morphology in the midcingulate cortex of Huntington's disease cases with predominant mood symptomatology. An increased proportion of activated microglia was observed in the midcingulate of all Huntington's disease cases and positively correlated with 1C2 burden. Alterations in the astrocytic glutamate transporter EAAT2 were observed in the midcingulate cortex of patients associated with mood symptoms. INTERPRETATION This study presents pathological changes in microglia and astrocytes in the midcingulate cortex in Huntington's disease, which coincide with mood symptom presentation. These findings further the understanding of neuroinflammation in Huntington's disease, a necessary step for developing inflammation-targeted therapeutics. ANN NEUROL 2023;94:895-910.
Collapse
Affiliation(s)
- Thulani Palpagama
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Aimee Rose Mills
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Mackenzie Wendy Ferguson
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | - Clinton Turner
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Lynette Tippett
- Centre for Brain Research and School of Psychology, Faculty of Sciences, University of Auckland, Auckland, New Zealand
| | - Bert van der Werf
- Department of Epidemiology and Biostatistics, Faculty of Medical and Health Sciences, School of Population Health, University of Auckland, Auckland, New Zealand
| | - Henry John Waldvogel
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard Lewis Maxwell Faull
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, 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, Auckland, New Zealand
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| |
Collapse
|
28
|
Xie M, Pallegar PN, Parusel S, Nguyen AT, Wu LJ. Regulation of cortical hyperexcitability in amyotrophic lateral sclerosis: focusing on glial mechanisms. Mol Neurodegener 2023; 18:75. [PMID: 37858176 PMCID: PMC10585818 DOI: 10.1186/s13024-023-00665-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the loss of both upper and lower motor neurons, resulting in muscle weakness, atrophy, paralysis, and eventually death. Motor cortical hyperexcitability is a common phenomenon observed at the presymptomatic stage of ALS. Both cell-autonomous (the intrinsic properties of motor neurons) and non-cell-autonomous mechanisms (cells other than motor neurons) are believed to contribute to cortical hyperexcitability. Decoding the pathological relevance of these dynamic changes in motor neurons and glial cells has remained a major challenge. This review summarizes the evidence of cortical hyperexcitability from both clinical and preclinical research, as well as the underlying mechanisms. We discuss the potential role of glial cells, particularly microglia, in regulating abnormal neuronal activity during the disease progression. Identifying early changes such as neuronal hyperexcitability in the motor system may provide new insights for earlier diagnosis of ALS and reveal novel targets to halt the disease progression.
Collapse
Affiliation(s)
- Manling Xie
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Praveen N Pallegar
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Sebastian Parusel
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
29
|
Bornstein R, Mulholland MT, Sedensky M, Morgan P, Johnson SC. Glutamine metabolism in diseases associated with mitochondrial dysfunction. Mol Cell Neurosci 2023; 126:103887. [PMID: 37586651 PMCID: PMC10773532 DOI: 10.1016/j.mcn.2023.103887] [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: 05/19/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023] Open
Abstract
Mitochondrial dysfunction can arise from genetic defects or environmental exposures and impact a wide range of biological processes. Among these are metabolic pathways involved in glutamine catabolism, anabolism, and glutamine-glutamate cycling. In recent years, altered glutamine metabolism has been found to play important roles in the pathologic consequences of mitochondrial dysfunction. Glutamine is a pleiotropic molecule, not only providing an alternate carbon source to glucose in certain conditions, but also playing unique roles in cellular communication in neurons and astrocytes. Glutamine consumption and catabolic flux can be significantly altered in settings of genetic mitochondrial defects or exposure to mitochondrial toxins, and alterations to glutamine metabolism appears to play a particularly significant role in neurodegenerative diseases. These include primary mitochondrial diseases like Leigh syndrome (subacute necrotizing encephalopathy) and MELAS (mitochondrial myopathy with encephalopathy, lactic acidosis, and stroke-like episodes), as well as complex age-related neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Pharmacologic interventions targeting glutamine metabolizing and catabolizing pathways appear to provide some benefits in cell and animal models of these diseases, indicating glutamine metabolism may be a clinically relevant target. In this review, we discuss glutamine metabolism, mitochondrial disease, the impact of mitochondrial dysfunction on glutamine metabolic processes, glutamine in neurodegeneration, and candidate targets for therapeutic intervention.
Collapse
Affiliation(s)
- Rebecca Bornstein
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, USA
| | - Michael T Mulholland
- Department of Applied Sciences, Translational Bioscience, Northumbria University, Newcastle, UK
| | - Margaret Sedensky
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, USA
| | - Phil Morgan
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, USA
| | - Simon C Johnson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA; Department of Neurology, University of Washington, Seattle, USA; Department of Applied Sciences, Translational Bioscience, Northumbria University, Newcastle, UK.
| |
Collapse
|
30
|
Li K, Zhu Z, Sun X, Zhao L, Liu Z, Xing J. Harnessing the therapeutic potential of mesenchymal stem cell-derived exosomes in cardiac arrest: Current advances and future perspectives. Biomed Pharmacother 2023; 165:115201. [PMID: 37480828 DOI: 10.1016/j.biopha.2023.115201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND Cardiac arrest (CA), characterized by sudden onset and high mortality rates, is one of the leading causes of death globally, with a survival rate of approximately 6-24%. Studies suggest that the restoration of spontaneous circulation (ROSC) hardly improved the mortality rate and prognosis of patients diagnosed with CA, largely due to ischemia-reperfusion injury. MAIN BODY Mesenchymal stem cells (MSCs) exhibit self-renewal and strong potential for multilineage differentiation. Their effects are largely mediated by extracellular vesicles (EVs). Exosomes are the most extensively studied subgroup of EVs. EVs mainly mediate intercellular communication by transferring vesicular proteins, lipids, nucleic acids, and other substances to regulate multiple processes, such as cytokine production, cell proliferation, apoptosis, and metabolism. Thus, exosomes exhibit significant potential for therapeutic application in wound repair, tissue reconstruction, inflammatory reaction, and ischemic diseases. CONCLUSION Based on similar pathological mechanisms underlying post-cardiac arrest syndrome involving various tissues and organs in many diseases, the review summarizes the therapeutic effects of MSC-derived exosomes and explores the prospects for their application in the treatment of CA.
Collapse
Affiliation(s)
- Ke Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun 130021, China.
| | - Zhu Zhu
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Xiumei Sun
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun 130021, China.
| | - Linhong Zhao
- Northeast Normal University, Changchun 130022, China.
| | - Zuolong Liu
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun 130021, China.
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
31
|
Corrigan M, O'Rourke A, Moran B, Fletcher J, Harkin A. Inflammation in the pathogenesis of depression: a disorder of neuroimmune origin. Neuronal Signal 2023; 7:NS20220054. [PMID: 37457896 PMCID: PMC10345431 DOI: 10.1042/ns20220054] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
There are several hypotheses concerning the underlying pathophysiological mechanisms of major depression, which centre largely around adaptive changes in neuronal transmission and plasticity, neurogenesis, and circuit and regional connectivity. The immune and endocrine systems are commonly implicated in driving these changes. An intricate interaction of stress hormones, innate immune cells and the actions of soluble mediators of immunity within the nervous system is described as being associated with the symptoms of depression. Bridging endocrine and immune processes to neurotransmission and signalling within key cortical and limbic brain circuits are critical to understanding depression as a disorder of neuroimmune origins. Emergent areas of research include a growing recognition of the adaptive immune system, advances in neuroimaging techniques and mechanistic insights gained from transgenic animals. Elucidation of glial-neuronal interactions is providing additional avenues into promising areas of research, the development of clinically relevant disease models and the discovery of novel therapies. This narrative review focuses on molecular and cellular mechanisms that are influenced by inflammation and stress. The aim of this review is to provide an overview of our current understanding of depression as a disorder of neuroimmune origin, focusing on neuroendocrine and neuroimmune dysregulation in depression pathophysiology. Advances in current understanding lie in pursuit of relevant biomarkers, as the potential of biomarker signatures to improve clinical outcomes is yet to be fully realised. Further investigations to expand biomarker panels including integration with neuroimaging, utilising individual symptoms to stratify patients into more homogenous subpopulations and targeting the immune system for new treatment approaches will help to address current unmet clinical need.
Collapse
Affiliation(s)
- Myles Corrigan
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Transpharmation Ireland, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Aoife M. O'Rourke
- School of Biochemistry and Immunology, Trinity Biosciences Institute, Trinity College, Dublin, Ireland
| | - Barry Moran
- School of Biochemistry and Immunology, Trinity Biosciences Institute, Trinity College, Dublin, Ireland
| | - Jean M. Fletcher
- School of Biochemistry and Immunology, Trinity Biosciences Institute, Trinity College, Dublin, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| |
Collapse
|
32
|
Fernández de la Torre M, Fiuza-Luces C, Laine-Menéndez S, Delmiro A, Arenas J, Martín MÁ, Lucia A, Morán M. Pathophysiology of Cerebellar Degeneration in Mitochondrial Disorders: Insights from the Harlequin Mouse. Int J Mol Sci 2023; 24:10973. [PMID: 37446148 DOI: 10.3390/ijms241310973] [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: 05/21/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
By means of a proteomic approach, we assessed the pathways involved in cerebellar neurodegeneration in a mouse model (Harlequin, Hq) of mitochondrial disorder. A differential proteomic profile study (iTRAQ) was performed in cerebellum homogenates of male Hq and wild-type (WT) mice 8 weeks after the onset of clear symptoms of ataxia in the Hq mice (aged 5.2 ± 0.2 and 5.3 ± 0.1 months for WT and Hq, respectively), followed by a biochemical validation of the most relevant changes. Additional groups of 2-, 3- and 6-month-old WT and Hq mice were analyzed to assess the disease progression on the proteins altered in the proteomic study. The proteomic analysis showed that beyond the expected deregulation of oxidative phosphorylation, the cerebellum of Hq mice showed a marked astroglial activation together with alterations in Ca2+ homeostasis and neurotransmission, with an up- and downregulation of GABAergic and glutamatergic neurotransmission, respectively, and the downregulation of cerebellar "long-term depression", a synaptic plasticity phenomenon that is a major player in the error-driven learning that occurs in the cerebellar cortex. Our study provides novel insights into the mechanisms associated with cerebellar degeneration in the Hq mouse model, including a complex deregulation of neuroinflammation, oxidative phosphorylation and glutamate, GABA and amino acids' metabolism.
Collapse
Affiliation(s)
- Miguel Fernández de la Torre
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
| | - Carmen Fiuza-Luces
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
| | - Sara Laine-Menéndez
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
| | - Aitor Delmiro
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
- Servicio de Bioquímica Clínica, Hospital Universitario "12 de Octubre", 28041 Madrid, Spain
| | - Joaquín Arenas
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
| | - Miguel Ángel Martín
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
- Servicio de Genética, Hospital Universitario "12 de Octubre", 28041 Madrid, Spain
| | - Alejandro Lucia
- Faculty of Sports Sciences, European University of Madrid, 28670 Madrid, Spain
- Spanish Network for Biomedical Research in Fragility and Healthy Aging (CIBERFES), 28029 Madrid, Spain
| | - María Morán
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
| |
Collapse
|
33
|
Ravache TT, Batistuzzo A, Nunes GG, Gomez TGB, Lorena FB, Do Nascimento BPP, Bernardi MM, Lima ERR, Martins DO, Campos ACP, Pagano RL, Ribeiro MO. Multisensory Stimulation Reverses Memory Impairment in Adrβ 3KO Male Mice. Int J Mol Sci 2023; 24:10522. [PMID: 37445699 DOI: 10.3390/ijms241310522] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Norepinephrine plays an important role in modulating memory through its beta-adrenergic receptors (Adrβ: β1, β2 and β3). Here, we hypothesized that multisensory stimulation would reverse memory impairment caused by the inactivation of Adrβ3 (Adrβ3KO) with consequent inhibition of sustained glial-mediated inflammation. To test this, 21- and 86-day-old Adrβ3KO mice were exposed to an 8-week multisensory stimulation (MS) protocol that comprised gustatory and olfactory stimuli of positive and negative valence; intellectual challenges to reach food; the use of hidden objects; and the presentation of food in ways that prompted foraging, which was followed by analysis of GFAP, Iba-1 and EAAT2 protein expression in the hippocampus (HC) and amygdala (AMY). The MS protocol reduced GFAP and Iba-1 expression in the HC of young mice but not in older mice. While this protocol restored memory impairment when applied to Adrβ3KO animals immediately after weaning, it had no effect when applied to adult animals. In fact, we observed that aging worsened the memory of Adrβ3KO mice. In the AMY of Adrβ3KO older mice, we observed an increase in GFAP and EAAT2 expression when compared to wild-type (WT) mice that MS was unable to reduce. These results suggest that a richer and more diverse environment helps to correct memory impairment when applied immediately after weaning in Adrβ3KO animals and indicates that the control of neuroinflammation mediates this response.
Collapse
Affiliation(s)
- Thaís T Ravache
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| | - Alice Batistuzzo
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| | - Gabriela G Nunes
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| | - Thiago G B Gomez
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| | - Fernanda B Lorena
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
- Departamento de Medicina Translacional, Universidade Federal de São Paulo 04023-062, SP, Brazil
| | - Bruna P P Do Nascimento
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
- Departamento de Medicina Translacional, Universidade Federal de São Paulo 04023-062, SP, Brazil
| | - Maria Martha Bernardi
- Graduate Program in Environmental and Experimental Pathology, Paulista University, São Paulo 04026-002, SP, Brazil
| | - Eduarda R R Lima
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo 01308-050, SP, Brazil
| | - Daniel O Martins
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo 01308-050, SP, Brazil
| | - Ana Carolina P Campos
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo 01308-050, SP, Brazil
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Rosana L Pagano
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo 01308-050, SP, Brazil
| | - Miriam O Ribeiro
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| |
Collapse
|
34
|
Bhatnagar A, Parmar V, Barbieri N, Bearoff F, Elefant F, Kortagere S. Novel EAAT2 activators improve motor and cognitive impairment in a transgenic model of Huntington's disease. Front Behav Neurosci 2023; 17:1176777. [PMID: 37351153 PMCID: PMC10282606 DOI: 10.3389/fnbeh.2023.1176777] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction Glutamate excitotoxicity is causal in striatal neurodegeneration underlying motor dysfunction and cognitive deficits in Huntington's disease (HD). Excitatory amino acid transporter 2 (EAAT2), the predominant glutamate transporter accounting for >90% of glutamate transport, plays a key role in preventing excitotoxicity by clearing excess glutamate from the intrasynaptic cleft. Accordingly, EAAT2 has emerged as a promising therapeutic target for prevention of neuronal excitotoxicity underlying HD and other neurodegenerative diseases. Methods We have previously designed novel EAAT2 positive allosteric modulator GT951, GTS467, and GTS551, with low nanomolar efficacy in glutamate uptake and favorable pharmacokinetic properties. In this study, we test the neuroprotective abilities of these novel EAAT2 activators in vivo using the robust Drosophila HD transgenic model expressing human huntingtin gene with expanded repeats (Htt128Q). Results All three compounds significantly restored motor function impaired under HD pathology over a wide dose range. Additionally, treatment with all three compounds significantly improved HD-associated olfactory associative learning and short-term memory defects, while GT951 and GTS551 also improved middle-term memory in low-performing group. Similarly, treatment with GT951 and GTS551 partially protected against early mortality observed in our HD model. Further, treatment with all three EAAT2 activators induced epigenetic expression of EAAT2 Drosophila homolog and several cognition-associated genes. Conclusion Together, these results highlight the efficacy of GT951, GTS467 and GTS551 in treating motor and cognitive impairments under HD pathology and support their development for treatment of HD.
Collapse
Affiliation(s)
- Akanksha Bhatnagar
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
| | - Visha Parmar
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Nicholas Barbieri
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Frank Bearoff
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Felice Elefant
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
35
|
Jo D, Lim YH, Jung YS, Kim YK, Song J. Circular RNA Tmcc1 improves astrocytic glutamate metabolism and spatial memory via NF-κB and CREB signaling in a bile duct ligation mouse model: transcriptional and cellular analyses. J Neuroinflammation 2023; 20:121. [PMID: 37217942 DOI: 10.1186/s12974-023-02806-w] [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: 01/04/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Hepatic encephalopathy-induced hyperammonemia alters astrocytic glutamate metabolism in the brain, which is involved in cognitive decline. To identify specific therapeutic strategies for the treatment of hepatic encephalopathy, various molecular signaling studies, such as non-coding RNA functional study, have been conducted. However, despite several reports of circular RNAs (circRNAs) in the brain, few studies of circRNAs in hepatic encephalopathy-induced neuropathophysiological diseases have been conducted. METHODS In this study, we performed RNA sequencing to identify whether the candidate circRNA cirTmcc1 is specifically expressed in the brain cortex in a bile duct ligation (BDL) mouse model of hepatic encephalopathy. RESULTS Based on transcriptional and cellular analysis, we investigated the circTmcc1-dysregulation-induced changes in the expression of several genes that are associated with intracellular metabolism and astrocyte function. We found that the circTmcc1 binds with the NF-κB p65-CREB transcriptional complex and regulates the expression of the astrocyte transporter EAAT2. Furthermore, circTmcc1 contributed to the secretion of proinflammatory mediators and glutamate metabolism in astrocytes and subsequently modulated an improvement in spatial memory by mediating neuronal synaptic plasticity. CONCLUSIONS Thus, circTmcc1 may be a promising circRNA candidate for targeted interventions to prevent and treat the neuropathophysiological complications that occur due to hepatic encephalopathy.
Collapse
Affiliation(s)
- Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Jeollanam-Do, Hwasun, 58128, Republic of Korea
- Chonnam National University, Seoyangro 264, Hwasun, 58128, Republic of Korea
| | - Yeong-Hwan Lim
- Department of Biochemistry, Chonnam National University Medical School, Seoyangro 264, Hwasun, 58128, Republic of Korea
| | - Yoon Seok Jung
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Jeollanam-Do, Hwasun, 58128, Republic of Korea
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Seoyangro 264, Hwasun, 58128, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Jeollanam-Do, Hwasun, 58128, Republic of Korea.
- Chonnam National University, Seoyangro 264, Hwasun, 58128, Republic of Korea.
| |
Collapse
|
36
|
Xu X, Wang J, Du S, Shen X, Lian J, Zhou J, Wang M, Feng W, Lv Z, Zhu J, Jin L, Sun H, Wu L, Wang X, Qiu H, Wang W, Teng H, Wang Y, Huang Z. Yes-associated protein regulates glutamate homeostasis through promoting the expression of excitatory amino acid transporter-2 in astrocytes via β-catenin signaling. Glia 2023; 71:1197-1216. [PMID: 36617748 DOI: 10.1002/glia.24332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 01/10/2023]
Abstract
The homeostasis of glutamate is mainly regulated by the excitatory amino acid transporters (EAATs), especially by EAAT2 in astrocytes. Excessive glutamate in the synaptic cleft caused by dysfunction or dysregulation of EAAT2 can lead to excitotoxicity, neuronal death and cognitive dysfunction. However, it remains unclear about the detailed regulation mechanism of expression and function of astrocytic EAAT2. In this study, first, we found increased neuronal death and impairment of cognitive function in YAPGFAP -CKO mice (conditionally knock out Yes-associated protein [YAP] in astrocytes), and identified EAAT2 as a downstream target of YAP through RNA sequencing. Second, the expression of EAAT2 was decreased in cultured YAP-/- astrocytes and the hippocampus of YAPGFAP -CKO mice, and glutamate uptake was reduced in YAP-/- astrocytes, but increased in YAP-upregulated astrocytes. Third, further investigation of the mechanism showed that the mRNA and protein levels of β-catenin were decreased in YAP-/- astrocytes and increased in YAP-upregulated astrocytes. Wnt3a activated YAP signaling and up-regulated EAAT2 through β-catenin. Furthermore, over-expression or activation of β-catenin partially restored the downregulation of EAAT2, the impairment of glutamate uptake, neuronal death and cognitive decline that caused by YAP deletion. Finally, activation of EAAT2 also rescued neuronal death and cognitive decline in YAPGFAP -CKO mice. Taken together, our study identifies an unrecognized role of YAP signaling in the regulation of glutamate homeostasis through the β-catenin/EAAT2 pathway in astrocytes, which may provide novel insights into the pathogenesis of brain diseases that closely related to the dysfunction or dysregulation of EAAT2, and promote the development of clinical strategy.
Collapse
Affiliation(s)
- Xingxing Xu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiaojiao Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Siyu Du
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xiya Shen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiashu Lian
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Zhou
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mianxian Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenjin Feng
- Zhejiang Sinogen Medical Equipment Co., Ltd., Wenzhou, China
| | - Zhaoting Lv
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Junzhe Zhu
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Lingting Jin
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huankun Sun
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Lihao Wu
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Xiaoning Wang
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Haoyu Qiu
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Honglin Teng
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Wang
- Clinical Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihui Huang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
37
|
Pajer K, Bellák T, Grósz T, Nógrádi B, Patai R, Sinkó J, Vinay L, Liabeuf S, Erdélyi M, Nógrádi A. Riluzole treatment modulates KCC2 and EAAT-2 receptor expression and Ca 2+ accumulation following ventral root avulsion injury. Eur J Cell Biol 2023; 102:151317. [PMID: 37099936 DOI: 10.1016/j.ejcb.2023.151317] [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: 07/26/2022] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023] Open
Abstract
Avulsion injury results in motoneuron death due to the increased excitotoxicity developing in the affected spinal segments. This study focused on possible short and long term molecular and receptor expression alterations which are thought to be linked to the excitotoxic events in the ventral horn with or without the anti-excitotoxic riluzole treatment. In our experimental model the left lumbar 4 and 5 (L4, 5) ventral roots of the spinal cord were avulsed. Treated animals received riluzole for 2 weeks. Riluzole is a compound that acts to block voltage-activated Na+ and Ca2+ channels. In control animals the L4, 5 ventral roots were avulsed without riluzole treatment. Expression of astrocytic EAAT-2 and that of KCC2 in motoneurons on the affected side of the L4 spinal segment were detected after the injury by confocal and dSTORM imaging, intracellular Ca2+ levels in motoneurons were quantified by electron microscopy. The KCC2 labeling in the lateral and ventrolateral parts of the L4 ventral horn was weaker compared with the medial part of L4 ventral horn in both groups. Riluzole treatment dramatically enhanced motoneuron survival but was not able to prevent the down-regulation of KCC2 expression in injured motoneurons. In contrast, riluzole successfully obviated the increase of intracellular calcium level and the decrease of EAAT-2 expression in astrocytes compared with untreated injured animals. We conclude that KCC2 may not be an essential component for survival of injured motoneurons and riluzole is able to modulate the intracellular level of calcium and expression of EAAT-2.
Collapse
Affiliation(s)
- Krisztián Pajer
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tamás Bellák
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tímea Grósz
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary; Department of Neurology, Albert Szent-Györgyi Health Center, University of Szeged, Szeged, Hungary
| | - Roland Patai
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - József Sinkó
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Laurent Vinay
- Institut de Neurosciences de la Timone, UMR 7289, CNRS and Aix Marseille Université, Campus Santé Timone, 13385 Marseille, France
| | - Sylvie Liabeuf
- Institut de Neurosciences de la Timone, UMR 7289, CNRS and Aix Marseille Université, Campus Santé Timone, 13385 Marseille, France
| | - Miklós Erdélyi
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
| |
Collapse
|
38
|
Fontana IC, Souza DG, Souza DO, Gee A, Zimmer ER, Bongarzone S. A Medicinal Chemistry Perspective on Excitatory Amino Acid Transporter 2 Dysfunction in Neurodegenerative Diseases. J Med Chem 2023; 66:2330-2346. [PMID: 36787643 PMCID: PMC9969404 DOI: 10.1021/acs.jmedchem.2c01572] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The excitatory amino acid transporter 2 (EAAT2) plays a key role in the clearance and recycling of glutamate - the major excitatory neurotransmitter in the mammalian brain. EAAT2 loss/dysfunction triggers a cascade of neurodegenerative events, comprising glutamatergic excitotoxicity and neuronal death. Nevertheless, our current knowledge regarding EAAT2 in neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD), is restricted to post-mortem analysis of brain tissue and experimental models. Thus, detecting EAAT2 in the living human brain might be crucial to improve diagnosis/therapy for ALS and AD. This perspective article describes the role of EAAT2 in physio/pathological processes and provides a structure-activity relationship of EAAT2-binders, bringing two perspectives: therapy (activators) and diagnosis (molecular imaging tools).
Collapse
Affiliation(s)
- Igor C Fontana
- School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom.,Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 - Neo floor seventh, 141 83 Stockholm, Sweden
| | - Débora G Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Av. Ipiranga, 6681 Porto Alegre, Brazil
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil
| | - Antony Gee
- School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Eduardo R Zimmer
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Av. Sarmento Leite 500, sala, 90035-003 Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry (PPGBioq), and Pharmacology and Therapeutics (PPGFT), Universidade Federal do Rio Grande do Sul, Av. Sarmento Leite 500, sala, 305 Porto Alegre, Brazil.,Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Av. Ipiranga, 6681 Porto Alegre, Brazil.,McGill University Research Centre for Studies in Aging, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Salvatore Bongarzone
- School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| |
Collapse
|
39
|
Xin J, Yang T, Wu X, Wu Y, Liu Y, Liu X, Jiang M, Gao W. Spatial transcriptomics analysis of zone-dependent hepatic ischemia-reperfusion injury murine model. Commun Biol 2023; 6:194. [PMID: 36804628 PMCID: PMC9938905 DOI: 10.1038/s42003-023-04564-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
Hepatic ischemia-reperfusion (I/R) injury is a common complication in liver transplantation. The connection between I/R-induced injury response and liver heterogeneity has yet to be fully understood. In this study, we converge histopathological examination with spatial transcriptomics to dissect I/R injury patterns and their associated molecular changes, which reveal that the pericentral zones are most sensitive to I/R injury in terms of histology, transcriptomic changes, and cell type dynamics. Bioinformatic analysis of I/R injury-related pathways predicts that celastrol can protect against liver I/R injury by inducing ischemic pre-conditioning, which is experimentally validated. Mechanistically, celastrol likely implements its protective effect against I/R injury by activating HIF1α signaling and represents a potential strategy for resolving liver I/R.
Collapse
Affiliation(s)
- Jiaqi Xin
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Ting Yang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiaoyi Wu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Yingting Wu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yi Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xuan Liu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Mengxi Jiang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Wei Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| |
Collapse
|
40
|
Hammad AM, Naser A, Amawi H, Hall FS, Tiwari AK, Al-Trad B. Effect of amoxicillin/clavulanic acid in attenuating pregabalin-induced condition place preference. Behav Brain Res 2023; 439:114244. [PMID: 36470419 DOI: 10.1016/j.bbr.2022.114244] [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: 06/10/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Substance abuse is a worldwide problem with serious repercussions for patients and the communities where they live. Pregabalin (Lyrica), is a medication commonly used to treat neuropathic pain. Like other analgesic medications there has been concern about pregabalin abuse and misuse. Although it was initially suggested that pregabalin, like other gabapentinoids, has limited abuse liability, questions still remain concerning this inquiry. Changes in glutamate system homeostasis are a hallmark of adaptations underlying drug dependence, including down-regulation of the glutamate transporter 1 (GLT-1; SLC1A2) and the cystine/glutamate antiporter (xCT; SLC7A11). In this study, it was found that pregabalin (90 mg/kg) produces a conditioned place preference (CPP), indicative of reinforcing effects that suggest a potential for abuse liability. Moreover, like other drugs of abuse, pregabalin also produced alterations in glutamate homeostasis, reducing the mRNA expression of Slc1a2 and Slc7a11 in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC). Amoxicillin clavulanic acid, a β-lactam antibiotic, blocked the reinforcing effects of pregabalin and normalized glutamate homeostasis. These results suggest that pregabalin has abuse potential that should be examined more critically, and that, moreover, the mechanisms underlying these effects are similar to those of other drugs of abuse, such as heroin and cocaine. Additionally, these results support previous findings showing normalization of glutamate homeostasis by β-lactam drugs that provides a novel potential therapeutic approach for the treatment of drug abuse and dependence.
Collapse
Affiliation(s)
- Alaa M Hammad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan.
| | - Asma'a Naser
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 21163, Jordan
| | - Haneen Amawi
- Department of Clinical Pharmacy and Pharmacy Practice, College of Pharmacy, Yarmouk University, Irbid 21163, Jordan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Bahaa Al-Trad
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 21163, Jordan
| |
Collapse
|
41
|
The Protective Role of Glutathione on Zinc-Induced Neuron Death after Brain Injuries. Int J Mol Sci 2023; 24:ijms24032950. [PMID: 36769273 PMCID: PMC9917832 DOI: 10.3390/ijms24032950] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Glutathione (GSH) is necessary for maintaining physiological antioxidant function, which is responsible for maintaining free radicals derived from reactive oxygen species at low levels and is associated with improved cognitive performance after brain injury. GSH is produced by the linkage of tripeptides that consist of glutamic acid, cysteine, and glycine. The adequate supplementation of GSH has neuroprotective effects in several brain injuries such as cerebral ischemia, hypoglycemia, and traumatic brain injury. Brain injuries produce an excess of reactive oxygen species through complex biochemical cascades, which exacerbates primary neuronal damage. GSH concentrations are known to be closely correlated with the activities of certain genes such as excitatory amino acid carrier 1 (EAAC1), glutamate transporter-associated protein 3-18 (Gtrap3-18), and zinc transporter 3 (ZnT3). Following brain-injury-induced oxidative stress, EAAC1 function is negatively impacted, which then reduces cysteine absorption and impairs neuronal GSH synthesis. In these circumstances, vesicular zinc is also released into the synaptic cleft and then translocated into postsynaptic neurons. The excessive influx of zinc inhibits glutathione reductase, which inhibits GSH's antioxidant functions in neurons, resulting in neuronal damage and ultimately in the impairment of cognitive function. Therefore, in this review, we explore the overall relationship between zinc and GSH in terms of oxidative stress and neuronal cell death. Furthermore, we seek to understand how the modulation of zinc can rescue brain-insult-induced neuronal death after ischemia, hypoglycemia, and traumatic brain injury.
Collapse
|
42
|
Sørensen NV, Benros ME. The Immune System and Depression: From Epidemiological to Clinical Evidence. Curr Top Behav Neurosci 2023; 61:15-34. [PMID: 35711028 DOI: 10.1007/7854_2022_369] [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: 06/15/2023]
Abstract
Depression is a frequent mental disorder with a substantial contribution to years lived with disability worldwide. In the search for new treatment targets, the immune system's contribution to the pathogenesis of depression has received increased attention as immune activation has been associated with depression in various epidemiological and case-control studies. Epidemiological studies have shown that immune exposures such as severe infections and autoimmune disorders increase the risk of depression. Furthermore, immune system activation has been indicated in case-control studies of depression revealing higher levels of key pro-inflammatory cytokines among patients with depression than healthy controls, particularly in blood and to some extent in the cerebrospinal fluid. Moreover, brain imaging studies indicate increased microglial activity during depression, and gut microbiota studies have documented alterations of gut microbiota composition to be associated with depression. Based on findings from animal and human studies, several immune-mediated molecular mechanisms have been suggested to underlie the association between increased immunological activity and depression. However, the research is challenged by the heterogeneity of the depression diagnosis and - to some extent - the precision of currently available technology for immune biomarker quantification, particularly regarding the assessment of low-grade neuroinflammation. Nonetheless, an enhanced understanding of the complex interactions between the immune system and the brain in the context of depression could pave the way for precision medicine approaches with immune-modulating treatment as a promising additional option in the treatment of depression.
Collapse
Affiliation(s)
- Nina Vindegaard Sørensen
- Biological and Precision Psychiatry, Copenhagen Research Centre for Mental Health, Mental Health Centre Copenhagen, Copenhagen University Hospital, Hellerup, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Eriksen Benros
- Biological and Precision Psychiatry, Copenhagen Research Centre for Mental Health, Mental Health Centre Copenhagen, Copenhagen University Hospital, Hellerup, Denmark.
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
43
|
Alijanpour S, Miryounesi M, Ghafouri-Fard S. The role of excitatory amino acid transporter 2 (EAAT2) in epilepsy and other neurological disorders. Metab Brain Dis 2023; 38:1-16. [PMID: 36173507 DOI: 10.1007/s11011-022-01091-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/15/2022] [Indexed: 02/03/2023]
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system (CNS). Excitatory amino acid transporters (EAATs) have important roles in the uptake of glutamate and termination of glutamatergic transmission. Up to now, five EAAT isoforms (EAAT1-5) have been identified in mammals. The main focus of this review is EAAT2. This protein has an important role in the pathoetiology of epilepsy. De novo dominant mutations, as well as inherited recessive mutation in this gene, have been associated with epilepsy. Moreover, dysregulation of this protein is implicated in a range of neurological diseases, namely amyotrophic lateral sclerosis, alzheimer's disease, parkinson's disease, schizophrenia, epilepsy, and autism. In this review, we summarize the role of EAAT2 in epilepsy and other neurological disorders, then provide an overview of the therapeutic modulation of this protein.
Collapse
Affiliation(s)
- Sahar Alijanpour
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Miryounesi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Arnst N, Redolfi N, Lia A, Bedetta M, Greotti E, Pizzo P. Mitochondrial Ca 2+ Signaling and Bioenergetics in Alzheimer's Disease. Biomedicines 2022; 10:3025. [PMID: 36551781 PMCID: PMC9775979 DOI: 10.3390/biomedicines10123025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is a hereditary and sporadic neurodegenerative illness defined by the gradual and cumulative loss of neurons in specific brain areas. The processes that cause AD are still under investigation and there are no available therapies to halt it. Current progress puts at the forefront the "calcium (Ca2+) hypothesis" as a key AD pathogenic pathway, impacting neuronal, astrocyte and microglial function. In this review, we focused on mitochondrial Ca2+ alterations in AD, their causes and bioenergetic consequences in neuronal and glial cells, summarizing the possible mechanisms linking detrimental mitochondrial Ca2+ signals to neuronal death in different experimental AD models.
Collapse
Affiliation(s)
- Nikita Arnst
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
| | - Nelly Redolfi
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
| | - Annamaria Lia
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
| | - Martina Bedetta
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
- Padova Neuroscience Center (PNC), University of Padova, 35131 Padua, Italy
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
- Study Centre for Neurodegeneration (CESNE), University of Padova, 35131 Padua, Italy
| |
Collapse
|
45
|
Temmermand R, Barrett JE, Fontana ACK. Glutamatergic systems in neuropathic pain and emerging non-opioid therapies. Pharmacol Res 2022; 185:106492. [PMID: 36228868 PMCID: PMC10413816 DOI: 10.1016/j.phrs.2022.106492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 01/14/2023]
Abstract
Neuropathic pain, a disease of the somatosensory nervous system, afflicts many individuals and adequate management with current pharmacotherapies remains elusive. The glutamatergic system of neurons, receptors and transporters are intimately involved in pain but, to date, there have been few drugs developed that therapeutically modulate this system. Glutamate transporters, or excitatory amino acid transporters (EAATs), remove excess glutamate around pain transmitting neurons to decrease nociception suggesting that the modulation of glutamate transporters may represent a novel approach to the treatment of pain. This review highlights and summarizes (1) the physiology of the glutamatergic system in neuropathic pain, (2) the preclinical evidence for dysregulation of glutamate transport in animal pain models, and (3) emerging novel therapies that modulate glutamate transporters. Successful drug discovery requires continuous focus on basic and translational methods to fully elucidate the etiologies of this disease to enable the development of targeted therapies. Increasing the efficacy of astrocytic EAATs may serve as a new way to successfully treat those suffering from this devastating disease.
Collapse
Affiliation(s)
- Rhea Temmermand
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - James E Barrett
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Andréia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| |
Collapse
|
46
|
Abram M, Jakubiec M, Reeb K, Cheng MH, Gedschold R, Rapacz A, Mogilski S, Socała K, Nieoczym D, Szafarz M, Latacz G, Szulczyk B, Kalinowska-Tłuścik J, Gawel K, Esguerra CV, Wyska E, Müller CE, Bahar I, Fontana ACK, Wlaź P, Kamiński RM, Kamiński K. Discovery of ( R)- N-Benzyl-2-(2,5-dioxopyrrolidin-1-yl)propanamide [ (R)-AS-1], a Novel Orally Bioavailable EAAT2 Modulator with Drug-like Properties and Potent Antiseizure Activity In Vivo. J Med Chem 2022; 65:11703-11725. [PMID: 35984707 PMCID: PMC9469208 DOI: 10.1021/acs.jmedchem.2c00534] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
(R)-7 [(R)-AS-1] showed broad-spectrum antiseizure activity across in vivo mouse seizure models: maximal electroshock (MES), 6 Hz (32/44 mA), acute pentylenetetrazol (PTZ), and PTZ-kindling. A remarkable separation between antiseizure activity and CNS-related adverse effects was also observed. In vitro studies with primary glia cultures and COS-7 cells expressing the glutamate transporter EAAT2 showed enhancement of glutamate uptake, revealing a stereoselective positive allosteric modulator (PAM) effect, further supported by molecular docking simulations. (R)-7 [(R)-AS-1] was not active in EAAT1 and EAAT3 assays and did not show significant off-target activity, including interactions with targets reported for marketed antiseizure drugs, indicative of a novel and unprecedented mechanism of action. Both in vivo pharmacokinetic and in vitro absorption, distribution, metabolism, excretion, toxicity (ADME-Tox) profiles confirmed the favorable drug-like potential of the compound. Thus, (R)-7 [(R)-AS-1] may be considered as the first-in-class small-molecule PAM of EAAT2 with potential for further preclinical and clinical development in epilepsy and possibly other CNS disorders.
Collapse
Affiliation(s)
- Michał Abram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688Krakow, Poland
| | - Marcin Jakubiec
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688Krakow, Poland
| | - Katelyn Reeb
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania19102, United States
| | - Mary Hongying Cheng
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15213, United States
| | - Robin Gedschold
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität Bonn, An der Immenburg 4, D-53121Bonn, Germany
| | - Anna Rapacz
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688Krakow, Poland
| | - Szczepan Mogilski
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688Krakow, Poland
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033Lublin, Poland
| | - Dorota Nieoczym
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033Lublin, Poland
| | - Małgorzata Szafarz
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688Krakow, Poland
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688Krakow, Poland
| | - Bartłomiej Szulczyk
- Department of Pharmacodynamics, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1B, 02-097Warsaw, Poland
| | - Justyna Kalinowska-Tłuścik
- Department of Crystal Chemistry and Crystal Physics, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387Krakow, Poland
| | - Kinga Gawel
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego 8B, 20-090Lublin, Poland
| | - Camila V Esguerra
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349Oslo, Norway
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688Krakow, Poland
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität Bonn, An der Immenburg 4, D-53121Bonn, Germany
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15213, United States
| | - Andréia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania19102, United States
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033Lublin, Poland
| | - Rafał M Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688Krakow, Poland
| | - Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688Krakow, Poland
| |
Collapse
|
47
|
Wood OWG, Yeung JHY, Faull RLM, Kwakowsky A. EAAT2 as a therapeutic research target in Alzheimer's disease: A systematic review. Front Neurosci 2022; 16:952096. [PMID: 36033606 PMCID: PMC9399514 DOI: 10.3389/fnins.2022.952096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/14/2022] [Indexed: 11/23/2022] Open
Abstract
Glutamate is the main excitatory neurotransmitter in the human central nervous system, responsible for a wide variety of normal physiological processes. Glutamatergic metabolism and its sequestration are tightly regulated in the normal human brain, and it has been demonstrated that dysregulation of the glutamatergic system can have wide-ranging effects both in acute brain injury and neurodegenerative diseases. The excitatory amino acid transporter 2 (EAAT2) is the dominant glutamatergic transporter in the human brain, responsible for efficient removal of glutamate from the synaptic cleft for recycling within glial cells. As such, it has a key role in maintaining excitatory-inhibitory homeostasis. Animal studies have demonstrated dysregulation or alterations of EAAT2 expression can have implications in neurodegenerative disorders. Despite extensive research into glutamatergic alterations in AD mouse models, there is a lack of studies examining the expression of EAAT2 within the AD human brain. In this systematic review, 29 articles were identified that either analyzed EAAT2 expression in the AD human brain or used a human-derived cell culture. Studies were inconclusive as to whether EAAT2 was upregulated or downregulated in AD. However, changes in localization and correlation between EAAT2 expression and symptomatology was noted. These findings implicate EAAT2 alterations as a key process in AD progression and highlight the need for further research into the characterization of EAAT2 processes in normal physiology and disease in human tissue and to identify compounds that can act as EAAT2 neuromodulators.
Collapse
Affiliation(s)
- Oliver W. G. Wood
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Jason H. Y. Yeung
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Richard L. M. Faull
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Pharmacology and Therapeutics, Galway Neuroscience Centre, School of Medicine, Ollscoil na Gaillimhe – University of Galway, Galway, Ireland
- *Correspondence: Andrea Kwakowsky
| |
Collapse
|
48
|
Sha L, Li G, Zhang X, Lin Y, Qiu Y, Deng Y, Zhu W, Xu Q. Pharmacological induction of AMFR increases functional EAAT2 oligomer levels and reduces epileptic seizures in mice. JCI Insight 2022; 7:160247. [PMID: 35938532 PMCID: PMC9462477 DOI: 10.1172/jci.insight.160247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
Dysregulation of excitatory amino acid transporter 2 (EAAT2) contributes to the development of temporal lobe epilepsy (TLE). Several strategies for increasing total EAAT2 levels have been proposed. However, the mechanism underlying the oligomeric assembly of EAAT2, impairment of which inhibits the formation of functional oligomers by EAAT2 monomers, is still poorly understood. In the present study, we identified E3 ubiquitin ligase AMFR as an EAAT2-interacting protein. AMFR specifically increased the level of EAAT2 oligomers rather than inducing protein degradation through K542-specific ubiquitination. By using tissues from humans with TLE and epilepsy model mice, we observed that AMFR and EAAT2 oligomer levels were simultaneously decreased in the hippocampus. Screening of 2386 FDA-approved drugs revealed that the most common analgesic/antipyretic medicine, acetaminophen (APAP), can induce AMFR transcriptional activation via transcription factor SP1. Administration of APAP protected against pentylenetetrazol-induced epileptogenesis. In mice with chronic epilepsy, APAP treatment partially reduced the occurrence of spontaneous seizures and greatly enhanced the antiepileptic effects of 17AAG, an Hsp90 inhibitor that upregulates total EAAT2 levels, when the 2 compounds were administered together. In summary, our studies reveal an essential role for AMFR in regulating the oligomeric state of EAAT2 and suggest that APAP can improve the efficacy of EAAT2-targeted antiepileptic treatments.
Collapse
Affiliation(s)
- Longze Sha
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Guanjun Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiuneng Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yarong Lin
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yunjie Qiu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yu Deng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wanwan Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
49
|
Raja A, Shekhar N, Singh H, Prakash A, Medhi B. In-silico discovery of dual active molecule to restore synaptic wiring against autism spectrum disorder via HDAC2 and H3R inhibition. PLoS One 2022; 17:e0268139. [PMID: 35877665 PMCID: PMC9312418 DOI: 10.1371/journal.pone.0268139] [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: 12/13/2021] [Accepted: 04/22/2022] [Indexed: 11/23/2022] Open
Abstract
Metal-dependent histone deacetylases (HDACs) are essential epigenetic regulators; their molecular and pharmacological roles in medically critical diseases such as neuropsychiatric disorders, neurodegeneration, and cancer are being studied globally. HDAC2’s differential expression in the central nervous system makes it an appealing therapeutic target for chronic neurological diseases like autism spectrum disorder. In this study, we identified H3R inhibitor molecules that are computationally effective at binding to the HDAC2 metal-coordinated binding site. The study highlights the importance of pitolisant in screening the potential H3R inhibitors by using a hybrid workflow of ligand and receptor-based drug discovery. The screened lead compounds with PubChem SIDs 103179850, 103185945, and 103362074 show viable binding with HDAC2 in silico. The importance of ligand contacts with the Zn2+ ion in the HDAC2 catalytic site is also discussed and investigated for a significant role in enzyme inhibition. The proposed H3R inhibitors 103179850, 103185945, and 103362074 are estimated as dual-active molecules to block the HDAC2-mediated deacetylation of the EAAT2 gene (SLC1A2) and H3R-mediated synaptic transmission irregularity and are, therefore, open for experimental validation.
Collapse
Affiliation(s)
- Anupam Raja
- Department of Pharmacology, PGIMER, Chandigarh, India
| | | | | | - Ajay Prakash
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, PGIMER, Chandigarh, India
- * E-mail:
| |
Collapse
|
50
|
Exosomes derived from bone marrow mesenchymal stem cells attenuate neurological damage in traumatic brain injury by alleviating glutamate-mediated excitotoxicity. Exp Neurol 2022; 357:114182. [PMID: 35901975 DOI: 10.1016/j.expneurol.2022.114182] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/08/2022] [Accepted: 07/21/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) is one of the major contributors to disability and death worldwide. Glutamate-mediated excitotoxicity, one of the secondary injuries occurring after TBI, leads to extreme neuronal apoptosis, and can be a potential target for intervention. Bone marrow mesenchymal stem cells-derived exosomes (BMSCs-Exos) have demonstrated neuroprotective effects on TBI. However, their precise role and the underlying mechanism by which they regulate glutamate-mediated excitotoxicity have not yet been determined. Therefore, this study aimed to determine whether BMSCs-Exos alleviate glutamate excitotoxicity post-TBI and their associated mechanism. METHODS BMSCs-Exos were extracted from the BMSCs incubation medium and identified by transmission electron microscopy, nanoparticle trafficking analysis, and western blotting. The neuroprotective effects of BMSCs-Exos on glutamate excitotoxicity were investigated in the glutamate-mediated excitotoxicity neuronal cell model and the TBI rat model (TBI induced by controlled cortical impact) using western blotting and TUNEL assay. Cortical lesion samples were collected post-TBI on day-1 and day-14 to study histology. In addition, cortical lesion volume on days 1, 3 and 7 following TBI was determined using T2-weighted magnetic resonance imaging (MRI), and cognitive function was assessed at 4 weeks following TBI using the Morris water maze (MWM) test. RESULTS BMSC-Exos were observed to be spherical with a mean diameter of 109.9 nm, and expressed exosomal markers CD9, CD81 and TSg101. BMSCs-Exos were efficiently endocytosed by astrocytes after co-incubation for 24 h. In vitro studies revealed that 125 μM of glutamate significantly induced neuronal apoptosis, which was attenuated by BMSCs-Exos in astrocyte-neuron co-cultures. This attenuation was mediated by the upregulation of glutamate transporter-1 (GLT-1) level and the downregulation of p-p38 MAPK level in astrocytes. Similar results were obtained in vivo, wherein we verified that PKH67-labeled BMSCs-Exos administered intravenously could reach the perilesional cortex crossing the blood-brain barrier and significantly reduce glutamate levels in the perilesional cortex of the TBI rat, accompanied by increased GLT-1 level and downregulation in p-p38 MAPK level. Additionally, western blotting and TUNEL staining also revealed that BMSCs-Exos significantly downregulated the expression of pro-apoptosis markers, including cleaved caspase-3 and cleaved caspase-9, and attenuated neuronal apoptosis following TBI. Immunohistochemical analysis and Nissl staining showed that BMSCs-Exos significantly increased GLT-1-positive cells, and the number of apoptotic neurons decreased in the perilesional cortex. Moreover, MRI and MWM results revealed that BMSCs-Exos significantly minimized cortical lesion volume and ameliorated cognitive function after TBI. The underlying neuroprotective mechanism of BMSCs-Exos may be due to an increase in GLT-1 level in astrocytes by blocking the p38 MAPK signaling pathway. CONCLUSION Taken together, our findings demonstrate that the implementation of BMSCs-Exos may be an effective prospective therapy for attenuating post-TBI neurological damage.
Collapse
|