1
|
Xu X, Xuan S, Chen S, Liu D, Xiao Q, Tu J. Increased excitatory amino acid transporter 2 levels in basolateral amygdala astrocytes mediate chronic stress-induced anxiety-like behavior. Neural Regen Res 2025; 20:1721-1734. [PMID: 39104111 PMCID: PMC11688569 DOI: 10.4103/nrr.nrr-d-23-01411] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/03/2024] [Accepted: 02/20/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202506000-00024/figure1/v/2024-08-05T133530Z/r/image-tiff The conventional perception of astrocytes as mere supportive cells within the brain has recently been called into question by empirical evidence, which has revealed their active involvement in regulating brain function and encoding behaviors associated with emotions. Specifically, astrocytes in the basolateral amygdala have been found to play a role in the modulation of anxiety-like behaviors triggered by chronic stress. Nevertheless, the precise molecular mechanisms by which basolateral amygdala astrocytes regulate chronic stress-induced anxiety-like behaviors remain to be fully elucidated. In this study, we found that in a mouse model of anxiety triggered by unpredictable chronic mild stress, the expression of excitatory amino acid transporter 2 was upregulated in the basolateral amygdala. Interestingly, our findings indicate that the targeted knockdown of excitatory amino acid transporter 2 specifically within the basolateral amygdala astrocytes was able to rescue the anxiety-like behavior in mice subjected to stress. Furthermore, we found that the overexpression of excitatory amino acid transporter 2 in the basolateral amygdala, whether achieved through intracranial administration of excitatory amino acid transporter 2 agonists or through injection of excitatory amino acid transporter 2-overexpressing viruses with GfaABC1D promoters, evoked anxiety-like behavior in mice. Our single-nucleus RNA sequencing analysis further confirmed that chronic stress induced an upregulation of excitatory amino acid transporter 2 specifically in astrocytes in the basolateral amygdala. Moreover, through in vivo calcium signal recordings, we found that the frequency of calcium activity in the basolateral amygdala of mice subjected to chronic stress was higher compared with normal mice. After knocking down the expression of excitatory amino acid transporter 2 in the basolateral amygdala, the frequency of calcium activity was not significantly increased, and anxiety-like behavior was obviously mitigated. Additionally, administration of an excitatory amino acid transporter 2 inhibitor in the basolateral amygdala yielded a notable reduction in anxiety level among mice subjected to stress. These results suggest that basolateral amygdala astrocytic excitatory amino acid transporter 2 plays a role in in the regulation of unpredictable chronic mild stress-induced anxiety-like behavior by impacting the activity of local glutamatergic neurons, and targeting excitatory amino acid transporter 2 in the basolateral amygdala holds therapeutic promise for addressing anxiety disorders.
Collapse
Affiliation(s)
- Xirong Xu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- University of Chinese of Academy of Sciences, Beijing, China
| | - Shoumin Xuan
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Shuai Chen
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- University of Chinese of Academy of Sciences, Beijing, China
| | - Dan Liu
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Qian Xiao
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Jie Tu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- University of Chinese of Academy of Sciences, Beijing, China
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
Collapse
|
2
|
Liu J, Sun K, Chen Q, Ding L. Advances in astrocytes in aneurysmal subarachnoid haemorrhage. Front Surg 2025; 12:1511242. [PMID: 40520688 PMCID: PMC12162669 DOI: 10.3389/fsurg.2025.1511242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 05/12/2025] [Indexed: 06/18/2025] Open
Abstract
Aneurysmal subarachnoid haemorrhage (aSAH) represents a subtype of stroke with a high incidence of morbidity and mortality. Astrocytes, the most abundant cell type in the central nervous system, play a pivotal role in brain injury and recovery following aSAH. They participate in synaptic remodelling, blood-brain barrier injury and the activation of the glial-mesenchymal-cervical lymphatic system. In this paper, we review the physiopathological functions and pathological changes of astrocytes after aSAH, exploring their mechanism of action in aSAH. We also summarise the evidence of therapeutic approaches to modulate astrocyte function after aSAH complications, and provide some new clues for future translational therapies to mitigate injury after aSAH.
Collapse
Affiliation(s)
| | | | | | - Lianshu Ding
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| |
Collapse
|
3
|
Qin Q, Zhang H, Li X, Ruan H, Liu S, Chen Y, Xu Z, Wang Y, Yan X, Jiang X. MiR-129-5p alleviates depression and anxiety by increasing astrocyte ATP production partly through targeting deubiquitinase Mysm1. PLoS One 2025; 20:e0322715. [PMID: 40344568 PMCID: PMC12064192 DOI: 10.1371/journal.pone.0322715] [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: 09/09/2024] [Accepted: 03/26/2025] [Indexed: 05/11/2025] Open
Abstract
Major depressive disorder (MDD) is a major global mental concern that severely affects quality of life, yet current pharmacological treatments remain limited in their effectiveness. Long-term chronic stress has been shown to increase the incidence of depression and anxiety. Micro RNAs (miRNAs) have been revealed to participate in the pathological process of depression and represent promising therapeutic targets. In this study, we found that microRNA-129-5p (miR-129-5p) was significantly decreased in the brains of depressive mice. Overexpression of miR-129-5p in the hippocampus effectively alleviated depressive-like behaviors and reduced the activation of microglial cells and astrocytes. In addition, ATP levels in depressive mice were significantly increased following miR-129-5p overexpression. The antidepressant effects of miR-129-5p were reversed when ATP function was blocked with the non-specific P2 receptor antagonist suramin. In vitro experiments revealed that miR-129-5p overexpression enhanced ATP production in astrocytes. Furthermore, using a dual-luciferase reporter assay, we found that miR-129-5p directly targeted Mysm1. When overexpressed in astrocytes, miR-129-5p significantly suppressed Mysm1 expression, promoted phosphorylation of p53 and AMPK, and enhanced the expression of PGC1α, factors previously associated with ATP production. Our findings highlight the crucial role of miR-129-5p in regulating depression, suggesting that miR-129-5p overexpression may serve as an effective strategy for antidepressant treatment.
Collapse
Affiliation(s)
- Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Heyang Zhang
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Xiaotong Li
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Huaqiang Ruan
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Shuirong Liu
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Yue Chen
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Zhenhua Xu
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Yan Wang
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Xinlong Yan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
- Anhui Medical University, Hefei, Anhui, China
- Jishou University, Jishou, Hunan, China
| |
Collapse
|
4
|
Alberquilla S, Nanclares C, Expósito S, Gall G, Kofuji P, Araque A, Martín ED, Moratalla R. Astrocytes Mediate Psychostimulant-Induced Alterations of Spike-Timing Dependent Synaptic Plasticity. Glia 2025; 73:1051-1067. [PMID: 39801264 PMCID: PMC11920680 DOI: 10.1002/glia.24672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 03/20/2025]
Abstract
At cellular and circuit levels, drug addiction is considered a dysregulation of synaptic plasticity. In addition, dysfunction of the glutamate transporter 1 (GLT-1) in the nucleus accumbens (NAc) has also been proposed as a mechanism underlying drug addiction. However, the cellular and synaptic impact of GLT-1 alterations in the NAc remain unclear. Here we show in the NAc that 10 days withdraw after 5 days treatment with cocaine or amphetamine decreases GLT-1 expression in astrocytes, which results in the prolongation of the excitatory postsynaptic potential (EPSP) decay kinetics in D1 receptor-containing medium spiny neurons (D1R-MSNs). Using the spike timing dependent plasticity (STDP) paradigm, we found that enlargement of EPSP duration results in switching the LTP elicited in control animals to LTD in psychostimulant-treated mice. In contrast to D1-MSNs, D2-MSNs did not display changes in EPSP kinetics and synaptic plasticity. Notably, the psychostimulant-induced synaptic transmission and synaptic plasticity effects were absent in IP3R2-/- mice, which lack astrocyte calcium signal, but were mimicked by the selective astrocytes stimulation with DREADDs. Finally, ceftriaxone, which upregulates GLT-1, restored normal GLT-1 function, EPSP kinetics, and synaptic plasticity in psychostimulant-treated mice. Therefore, we propose that cocaine and amphetamine increase dopaminergic levels in the NAc, which stimulates astrocytes and downregulates the GLT-1. The decreased GLT-1 function prolonged the EPSP kinetics, leading to the modulation of the STDP, transforming the LTP observed in control animals into LTD in psychostimulant-treated mice. Present work reveals a novel mechanism underlying the synaptic plasticity changes induced by these drugs of abuse.
Collapse
Affiliation(s)
- Samuel Alberquilla
- Cajal Institute, CSICMadridSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
| | - Carmen Nanclares
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Grace Gall
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Paulo Kofuji
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Alfonso Araque
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Rosario Moratalla
- Cajal Institute, CSICMadridSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
5
|
Zhang X, Ren W, Li D, Si Y, Yang C, Wang L, Song L. An excitatory amino acid transporter 1 acts as a novel glutamine transporter and immune modulator in the oyster Crassostrea gigas. Int J Biol Macromol 2025; 310:143127. [PMID: 40228773 DOI: 10.1016/j.ijbiomac.2025.143127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Glutamine (Gln) plays a pivotal role in immunoregulation in vertebrates through transporter-mediated transport, whereas mechanisms in invertebrates remain largely unexplored. In this study, an excitatory amino acid transporter 1 (designated CgEAAT1) was identified from the Pacific oyster Crassostrea gigas, which is capable of transporting both Gln and glutamate (Glu), a functional characteristic distinct from vertebrate EAAT1 homologs. CgEAAT1 harbors a conserved Gltp domain and exhibits structural homology with vertebrate SLC1A5. Gln incubation significantly elevated CgEAAT1 and CgIL17-1 expression in hemocytes, concomitant with increased granulocyte proportions. Silencing of CgEAAT1 led to a substantial decrease in intracellular Gln levels, CgIL17-1 expression, and granulocyte proportion. Fluorescence-based assays confirmed attenuated Gln uptake following CgEAAT1 knockdown. Inhibition of CgEAAT1 expression and activity induced significant reductions in intracellular Gln concentrations and CgIL17-1 transcript levels within 6 h, with effects attenuating by 12 h, indicating complex Gln transport mechanisms in oysters. Exogenous Gln supplementation resulted in lowered larval mortality following Vibrio splendidus challenge, whereas CgEAAT1 inhibition increased mortality. These findings demonstrate that CgEAAT1 regulates hemocyte immune function through Gln transport, enhancing pathogen resistance via promotion of IL17-1 expression and granulocyte production. This research provides novel insights into Gln transport and immune regulatory mechanisms underlying molluscan environmental adaptation.
Collapse
Affiliation(s)
- Xueshu Zhang
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Wenjing Ren
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Deliang Li
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Yiran Si
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Chuanyan Yang
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
6
|
Zuschlag Y, Pommereau A, Warkentin J, Licher T, Bärenz F. The development of a high-throughput acoustic droplet ejection mass-spectrometry assay and a solid-supported membrane (SSM)-based electrophysiological assay to study the pharmacological inhibition of SLC1-A3, -A2 and -A1 in a drug discovery program. Front Pharmacol 2025; 16:1544682. [PMID: 40308755 PMCID: PMC12041765 DOI: 10.3389/fphar.2025.1544682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction The solute carrier (SLC) family comprises a diverse group of membrane proteins essential for transporting a variety of substrates across cellular membranes. These transporters play crucial roles in cellular homeostasis, nutrient uptake, and neurotransmitter clearance. The SLC1 subfamily, specifically SLC1A3 (EAAT1), SLC1A2 (EAAT2), and SLC1A1 (EAAT3), are excitatory amino acid transporters that regulate glutamate concentrations in the synaptic cleft, making them important targets for neurological disorder therapeutics. Despite their significance, drug discovery efforts targeting these transporters have been hampered by limitations in available screening methodologies. Methods We are utilizing advanced methodologies such as Acoustic Droplet Ejection Mass Spectrometry (ADE-MS) and Solid Supported Membrane (SSM)-based electrophysiology to develop assays for the SLC1 family members: SLC1A3 (EAAT1), SLC1A2 (EAAT2), and SLC1A1 (EAAT3). Results and Discussion In this manuscript, we present the successful development of novel assays specifically designed for drug discovery applications targeting the SLC1 family members. Our Acoustic Droplet Ejection Mass Spectrometry (ADE-MS) platform demonstrated high sensitivity and reproducibility in detecting substrate transport activity across all three transporters. The complementary Solid Supported Membrane (SSM)-based electrophysiology assay provided real-time kinetic measurements of transporter function with minimal background interference. These assays exhibited Z' factors exceeding 0.7, indicating their robustness for high-throughput screening campaigns. Initial validation using known inhibitors confirmed the assays' ability to identify compounds with varying potencies and mechanisms of action against SLC1A3, SLC1A2, and SLC1A1. Conclusion We endeavor to establish robust assays that can facilitate future drug discovery campaigns.
Collapse
Affiliation(s)
| | | | | | | | - Felix Bärenz
- Sanofi, Integrated Drug Discovery, Frankfurt, Germany
| |
Collapse
|
7
|
Faustmann TJ, Corvace F, Faustmann PM, Ismail FS. Influence of antipsychotic drugs on microglia-mediated neuroinflammation in schizophrenia: perspectives in an astrocyte-microglia co-culture model. Front Psychiatry 2025; 16:1522128. [PMID: 40171306 PMCID: PMC11959008 DOI: 10.3389/fpsyt.2025.1522128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/12/2025] [Indexed: 04/03/2025] Open
Abstract
Schizophrenia is a severe mental disorder with a strong lifetime impact on patients' health and wellbeing. Usually, symptomatic treatment includes typical or atypical antipsychotics. Study findings show an involvement of low-grade inflammation (blood, brain parenchyma, and cerebrospinal fluid) in schizophrenia. Moreover, experimental and neuropathological evidence suggests that reactive microglia, which are the main resident immune cells of the central nervous system (CNS), have a negative impact on the differentiation and function of oligodendrocytes, glial progenitor cells, and astrocytes, which results in the disruption of neuronal networks and dysregulated synaptic transmission, contributing to the pathophysiology of schizophrenia. Here, the role of microglial cells related to neuroinflammation in schizophrenia was discussed to be essential. This review aims to summarize the evidence for the influence of antipsychotics on microglial inflammatory mechanisms in schizophrenia. Furthermore, we propose an established astrocyte-microglia co-culture model for testing regulatory mechanisms and examining the effects of antipsychotics on glia-mediated neuroinflammation. This could lead to a better understanding of how typical and atypical antipsychotics can be used to address positive and negative symptoms in schizophrenia and comorbidities like inflammatory diseases or the status of low-grade inflammation.
Collapse
Affiliation(s)
- Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Franco Corvace
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Pedro M. Faustmann
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Fatme Seval Ismail
- Department of Neurology, Klinikum Vest, Academic Teaching Hospital of the Ruhr University Bochum, Recklinghausen, Germany
| |
Collapse
|
8
|
Gupta S, Gupta AK, Mehan S, Khan Z, Gupta GD, Narula AS. Disruptions in cellular communication: Molecular interplay between glutamate/NMDA signalling and MAPK pathways in neurological disorders. Neuroscience 2025; 569:331-353. [PMID: 39809360 DOI: 10.1016/j.neuroscience.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/30/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Neurological disorders significantly impact the central nervous system, contributing to a growing public health crisis globally. The spectrum of these disorders includes neurodevelopmental and neurodegenerative diseases. This manuscript reviews the crucial roles of cellular signalling pathways in the pathophysiology of these conditions, focusing primarily on glutaminase/glutamate/NMDA receptor signalling, alongside the mitogen-activated protein kinase (MAPK) pathways-ERK1/2, C-JNK, and P38 MAPK. Activation of these pathways is often correlated with neuronal excitotoxicity, apoptosis, and inflammation, leading to many other pathological conditions such as traumatic brain injury, stroke, and brain tumor. The interplay between glutamate overstimulation and MAPK signalling exacerbates neurodegenerative processes, underscoring the complexity of cellular communication in maintaining neuronal health. Dysfunctional signalling alters synaptic plasticity and neuronal survival, contributing to cognitive impairments in various neurological diseases. The manuscript emphasizes the potential of targeting these signalling pathways for therapeutic interventions, promoting neuroprotection and reducing neuroinflammation. Incorporating insights from precision medicine and innovative drug delivery systems could enhance treatment efficacy. Overall, understanding the intricate mechanisms of these pathways is essential for developing effective strategies to mitigate the impact of neurological disorders and improve patient outcomes. This review highlights the necessity for further exploration into these signalling cascades to facilitate advancements in therapeutic approaches, ensuring better prognoses for individuals affected by neurological conditions.
Collapse
Affiliation(s)
- Sumedha Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Abhishek Kumar Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India. https://mehanneuroscience.org
| | - Zuber Khan
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
9
|
McManus RM, Komes MP, Griep A, Santarelli F, Schwartz S, Ramón Perea J, Schlachetzki JCM, Bouvier DS, Khalil MA, Lauterbach MA, Heinemann L, Schlüter T, Pour MS, Lovotti M, Stahl R, Duthie F, Rodríguez-Alcázar JF, Schmidt SV, Spitzer J, Noori P, Maillo A, Boettcher A, Herron B, McConville J, Gomez-Cabrero D, Tegnér J, Glass CK, Hiller K, Latz E, Heneka MT. NLRP3-mediated glutaminolysis controls microglial phagocytosis to promote Alzheimer's disease progression. Immunity 2025; 58:326-343.e11. [PMID: 39904338 DOI: 10.1016/j.immuni.2025.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/14/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
Activation of the NLRP3 inflammasome has been implicated in the pathogenesis of Alzheimer's disease (AD) via the release of IL-1β and ASC specks. However, whether NLRP3 is involved in pathways beyond this remained unknown. Here, we found that Aβ deposition in vivo directly triggered NLRP3 activation in APP/PS1 mice, which model many features of AD. Loss of NLRP3 increased glutamine- and glutamate-related metabolism and increased expression of microglial Slc1a3, which was associated with enhanced mitochondrial and metabolic activity. The generation of α-ketoglutarate during this process impacted cellular function, including increased clearance of Aβ peptides as well as epigenetic and gene transcription changes. This pathway was conserved between murine and human cells. Critically, we could mimic this effect pharmacologically using NLRP3-specific inhibitors, but only with chronic NLRP3 inhibition. Together, these data demonstrate an additional role for NLRP3, where it can modulate mitochondrial and metabolic function, with important downstream consequences for the progression of AD.
Collapse
Affiliation(s)
- Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Max P Komes
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Angelika Griep
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Francesco Santarelli
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | | | - Juan Ramón Perea
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid 28049, Spain
| | - Johannes C M Schlachetzki
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - David S Bouvier
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4367 Belvaux, Luxembourg; Laboratoire National de Santé (LNS), National Center of Pathology (NCP), Dudelange, Luxembourg; Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
| | - Michelle-Amirah Khalil
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Mario A Lauterbach
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Lea Heinemann
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Titus Schlüter
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Mehran Shaban Pour
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Marta Lovotti
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Rainer Stahl
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Fraser Duthie
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | | | - Susanne V Schmidt
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Jasper Spitzer
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Peri Noori
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Alberto Maillo
- Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain
| | - Andreas Boettcher
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Brian Herron
- Regional Neuropathology Service, Institute of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust, Grosvenor Road, Belfast BT12 6BL, Northern Ireland
| | | | - David Gomez-Cabrero
- Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Jesper Tegnér
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia; Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia; Science for Life Laboratory, Tomtebodavagen 23A, 17165, Solna, Sweden
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Deutsches Rheuma-Forschungszentrum (DRFZ), Charitéplatz 1, 10117 Berlin, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany; Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4367 Belvaux, Luxembourg; Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
10
|
Bernardo HT, Lodetti G, de Farias ACS, de Pieri Pickler K, Baldin SL, Dondossola ER, Rico EP. Naltrexone Alters Neurochemical and Behavioral Parameters in a Zebrafish Model of Repeated Alcohol Exposure. Neurochem Res 2025; 50:97. [PMID: 39920352 DOI: 10.1007/s11064-025-04349-3] [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: 10/01/2024] [Revised: 01/04/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
Between the neurotransmission systems modulated by alcohol, the opioid system has been receiving attention in studies that seek to understand its relationship to the effects of addictive substances and different neuropsychiatric disorders. The use of naltrexone stands out in determining the mechanisms of the opioid system, as it acts as an opioid antagonist and consequently generates neurochemical responses. This study aimed to evaluate the pharmacological modulation of opioids on behavioral and neurobiological aspects in adult zebrafish submitted to the protocol of repeated exposure to ethanol and treated with naltrexone. Opioid modulation using naltrexone has been shown to modulate anxiety-like behavior, presenting anxiolytic properties in isolation, in addition to reversing the anxiogenic effect of ethanol through the Novel tank and Light/dark test. Naltrexone increased serotonin and dopamine levels, while ethanol antagonized these effects. In contrast, the interaction between ethanol and naltrexone raised noradrenaline levels. Naltrexone altered glutamate levels, however, ethanol reversed it. Ethanol acted on glutamate transporters increasing their activities, while naltrexone treatment reduced activities. No significant results were found in the pro-oxidant parameters, however, ethanol reduced SOD activity while naltrexone reversed. The same occurred in CAT activity. Also, naltrexone up-regulated the expression of genes related to the dopaminergic, glutamatergic, and opioid systems. The genes used as markers of the inflammatory process and glial activity were modulated by ethanol and together with naltrexone, respectively. Taken together, our findings reinforce the importance of opioid signaling on biochemical and molecular bases related to neuropsychiatric behaviors and diseases, such as anxiety and substance dependence.
Collapse
Affiliation(s)
- Henrique Teza Bernardo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Guilherme Lodetti
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Ana Caroline Salvador de Farias
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Karolyne de Pieri Pickler
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Samira Leila Baldin
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Eduardo Ronconi Dondossola
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Eduardo Pacheco Rico
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
11
|
Chen J, Liu C, Li L, Tao W, Zhang X, Zhao S, Wang C, Huang L. Exogenous leptin alleviates glutamate-excitotoxic injury caused by cerebral ischemia-reperfusion in mice by affecting the expression of glutamate transporters. Brain Res 2024; 1845:149201. [PMID: 39197570 DOI: 10.1016/j.brainres.2024.149201] [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: 06/09/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Ischemic stroke is characterized by high morbidity and mortality and a lack of effective therapeutic interventions. Leptin plays an important role in regulating oxidative stress, angiogenesis, hematopoiesis, etc. Although recent studies have found a neuroprotective effect of leptin, little is known about its role in cerebral ischemia. This study explores the possible roles and potential preventative mechanisms of leptin in cerebral ischemia-reperfusion injury (CIRI). An in vivo middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model was used to replicate the CIRI model, low (0.5 mg/kg), medium (1 mg/kg) and high (2 mg/kg) concentrations of leptin were injected intraperitoneally immediately after inserting the embolic line. After 1.5 h of ischemia and 24 h of reperfusion, we examined the neural function of the mice, collected brain tissue for histological examination, and screened for the optimal concentrations of leptin intervention. On this basis, we observed the changes of cortical apoptosis injury, intracellular calcium fluorescence intensity and astrocyte glial fibrillary acidic protein (GFAP) expression and morphological changes. In addition, we also tested the expression of transporters and metabolism-related enzymes (VGLUT-1, VGLUT-2, GLAST, GLT-1, GS, ATP1α1), the expression of inflammatory factors and the content of glutamate (Glu). Compared with the I/R group, we found that leptin improved neurological deficits, reduced the area of infarcts, maintained the normal morphology of astrocytes (AST), downregulated the expression of VGLUT-1, and upregulates the expression of GLT-1 and GLAST, thereby reducing the content of Glu in the synaptic cleft. Our studies suggest that leptin may have a neuroprotective effect by decreasing the excitotoxicity of glutamate.
Collapse
Affiliation(s)
- Jie Chen
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Chenxu Liu
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Li Li
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - WeiTing Tao
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Xiaonan Zhang
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Shidi Zhao
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Chun Wang
- Department of General Medicine, Second Affiliated Hospital of Bengbu Medical University, Bengbu Anhui 233040, China.
| | - Li Huang
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China.
| |
Collapse
|
12
|
Shang S, Wang L, Lu X. β-Hydroxybutyrate enhances astrocyte glutamate uptake through EAAT1 expression regulation. Mol Cell Neurosci 2024; 131:103959. [PMID: 39179164 DOI: 10.1016/j.mcn.2024.103959] [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: 04/07/2024] [Revised: 07/28/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024] Open
Abstract
β-Hydroxybutyrate (BHB) has been reported to exert neuroprotective functions and is considered a promising treatment for neurodegenerative diseases such as Parkinson's and Alzheimer's. Numerous studies have revealed BHB's multifaceted roles, including anti-senescence, anti-oxidative, and anti-inflammatory activities. However, the underlying mechanisms warrant further investigation. Astrocytes, the most abundant glial cells in the central nervous system, play a pivotal role in the development and progression of neurodegenerative diseases. While BHB is known to alter neuronal metabolism and function, its effects on astrocytes remain poorly understood. In this study, we conducted transcriptome sequencing analysis to identify differentially expressed genes induced by BHB in astrocytes and found that the gene Solute carrier family 1 member 3 (Slc1a3), encoding the glutamate transporter EAAT1, was significantly upregulated by BHB treatment. Cellular and animal-based experiments confirmed an increase in EAAT1 protein expression in primary astrocytes and the hippocampus of mice treated with BHB. This upregulation may be due to the activation of the Ca2+/CAMKII pathway by BHB. Furthermore, BHB improved astrocytes' glutamate uptake and partially restored neuronal viability impaired by glutamate-induced excitotoxicity when astrocytes were functionalized. Our results suggest that BHB may alleviate neuronal damage caused by excessive glutamate by enhancing the glutamate absorption and uptake capacity of astrocytes. This study proposes a novel mechanism for the neuroprotective effects of BHB and reinforces its beneficial impact on the central nervous system (CNS).
Collapse
Affiliation(s)
- Sen Shang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, PR China
| | - Leilei Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, PR China
| | - Xiaoyun Lu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, PR China.
| |
Collapse
|
13
|
Molina SJ, Corsi GN, Araujo Añon LC, Guelman LR. Sex-dependent effects of short-term ethanol, energy drinks and acute noise exposure on hippocampal oxidative balance and glutamate transporter EAAT-1 during rat adolescence. Neurotoxicology 2024; 105:147-157. [PMID: 39366467 DOI: 10.1016/j.neuro.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/17/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
It is known that human adolescents often consume ethanol (EtOH) alone or mixed with energy drinks (ED), especially in noisy environments. Although these agents impact the developing brain, their effects after brief exposure or when presented together remain unclear. Given that few animal studies in this subject are available, this research aimed to study the effects of a brief exposure to these stimuli on the oxidative state and EAAT-1 glutamate transporter levels in the developing rat hippocampus (HC). Adolescent Wistar rats were subjected to a two-bottle choice, limited access to drinking in the dark paradigm, for EtOH and EtOH+ED intake, for 4 days, and subsequent acute noise exposure. Next, hippocampal catalase activity, reactive oxygen species (ROS), glutaredoxin-1 (Grx-1) and glutamate transporter EAAT-1 levels were assessed. Results showed sex-dependent alterations after exposure to these stimuli: Females consuming EtOH had higher hippocampal ROS levels, which decreased when combined with noise; males showed reduced ROS levels only after noise exposure. No significant changes occurred in catalase activity, Grx-1, or EAAT-1 levels with EtOH and noise exposure in neither sex. Additionally, ED raised EtOH consumption in both sexes, normalizing ROS levels only in females when combined with EtOH. Finally, ED consumption altered Grx-1 and EAAT-1 levels in both sexes. In summary, brief exposure to these stimuli induced sex-dependent alterations, suggesting differentiated coping strategies between sexes. Whereas ED consumption may have antioxidant effects in some cases, it could also increase excitotoxicity risk. These novel findings raise questions for future research on the underlying corresponding mechanisms.
Collapse
Affiliation(s)
- Sonia Jazmín Molina
- Universidad de Buenos Aires. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBA-CONICET). Facultad de Medicina, Buenos Aires, Argentina.
| | - Gonzalo Nahuel Corsi
- Universidad de Buenos Aires. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBA-CONICET). Facultad de Medicina, Buenos Aires, Argentina
| | - Lara Candela Araujo Añon
- Universidad de Buenos Aires. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBA-CONICET). Facultad de Medicina, Buenos Aires, Argentina
| | - Laura Ruth Guelman
- Universidad de Buenos Aires. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBA-CONICET). Facultad de Medicina, Buenos Aires, Argentina; Universidad de Buenos Aires. Facultad de Medicina, 1ª Cátedra de Farmacología, Buenos Aires, Argentina
| |
Collapse
|
14
|
Karagöl A, Karagöl T, Li M, Zhang S. Inhibitory Potential of the Truncated Isoforms on Glutamate Transporter Oligomerization Identified by Computational Analysis of Gene-Centric Isoform Maps. Pharm Res 2024; 41:2173-2187. [PMID: 39487385 PMCID: PMC11599315 DOI: 10.1007/s11095-024-03786-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVE Glutamate transporters play a key role in central nervous system physiology by maintaining excitatory neurotransmitter homeostasis. Biological assemblies of the transporters, consisting of cyclic homotrimers, emerge as a crucial aspect of glutamate transporter modulation. Hence targeting heteromerization promises an effective approach for modulator design. On the other hand, the dynamic nature of transcription allows for the generation of transporter isoforms in structurally distinct manners. METHODS The potential isoforms were identified through the analysis of computationally generated gene-centric isoform maps. The conserved features of isoform sequences were revealed by computational chemistry methods and subsequent structural analysis of AlphaFold2 predictions. Truncated isoforms were further subjected to a wide range of docking analyses, 50ns molecular dynamics simulations, and evolutionary coupling analyses. RESULTS Energetic landscapes of isoform-canonical transporter complexes suggested an inhibitory potential of truncated isoforms on glutamate transporter bio-assembly. Moreover, isoforms that mimic the trimerization domain (in particular, TM2 helices) exhibited stronger interactions with canonical transporters, underscoring the role of transmembrane helices in isoform interactions. Additionally, self-assembly dynamics observed in truncated isoforms mimicking canonical TM5 helices indicate a potential protective role against unwanted interactions with canonical transporters. CONCLUSION Our computational studies on glutamate transporters offer insights into the roles of alternative splicing on protein interactions and identifies potential drug targets for physiological or pathological processes.
Collapse
Affiliation(s)
- Alper Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Taner Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Mengke Li
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
15
|
Yang XM, Yu H, Li JX, Li N, Li C, Xu DH, Zhang H, Fang TH, Wang SJ, Yan PY, Han BB. Excitotoxic Storms of Ischemic Stroke: A Non-neuronal Perspective. Mol Neurobiol 2024; 61:9562-9581. [PMID: 38662299 DOI: 10.1007/s12035-024-04184-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
Numerous neurological disorders share a fatal pathologic process known as glutamate excitotoxicity. Among which, ischemic stroke is the major cause of mortality and disability worldwide. For a long time, the main idea of developing anti-excitotoxic neuroprotective agents was to block glutamate receptors. Despite this, there has been little successful clinical translation to date. After decades of "neuron-centered" views, a growing number of studies have recently revealed the importance of non-neuronal cells. Glial cells, cerebral microvascular endothelial cells, blood cells, and so forth are extensively engaged in glutamate synthesis, release, reuptake, and metabolism. They also express functional glutamate receptors and can listen and respond for fast synaptic transmission. This broadens the thoughts of developing excitotoxicity antagonists. In this review, the critical contribution of non-neuronal cells in glutamate excitotoxicity during ischemic stroke will be emphasized in detail, and the latest research progress as well as corresponding therapeutic strategies will be updated at length, aiming to reconceptualize glutamate excitotoxicity in a non-neuronal perspective.
Collapse
Affiliation(s)
- Xiao-Man Yang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, People's Republic of China
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Hao Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Jia-Xin Li
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, People's Republic of China
| | - Na Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Chong Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Dong-Han Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, People's Republic of China
| | - Hao Zhang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, People's Republic of China
| | - Tian-He Fang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Shi-Jun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.
| | - Pei-Yu Yan
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, People's Republic of China.
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, People's Republic of China.
- Zhuhai MUST Science and Technology Research Institute, Macau University of Science and Technology, Macau, People's Republic of China.
| | - Bing-Bing Han
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.
| |
Collapse
|
16
|
Feng Y, Sun L, Dang X, Liu D, Liao Z, Yao J, Zhang Y, Deng Z, Li J, Zhao M, Liu F. Aberrant glycosylation in schizophrenia: insights into pathophysiological mechanisms and therapeutic potentials. Front Pharmacol 2024; 15:1457811. [PMID: 39286629 PMCID: PMC11402814 DOI: 10.3389/fphar.2024.1457811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
Schizophrenia (SCZ) is a severe neuropsychiatric disorder characterized by cognitive, affective, and social dysfunction, resulting in hallucinations, delusions, emotional blunting, and disordered thinking. In recent years, proteomics has been increasingly influential in SCZ research. Glycosylation, a key post-translational modification, can alter neuronal stability and normal signaling in the nervous system by affecting protein folding, stability, and cellular signaling. Recent research evidence suggests that abnormal glycosylation patterns exist in different brain regions in autopsy samples from SCZ patients, and that there are significant differences in various glycosylation modification types and glycosylation modifying enzymes. Therefore, this review explores the mechanisms of aberrant modifications of N-glycosylation, O-glycosylation, glycosyltransferases, and polysialic acid in the brains of SCZ patients, emphasizing their roles in neurotransmitter receptor function, synaptic plasticity, and neural adhesion. Additionally, the effects of antipsychotic drugs on glycosylation processes and the potential for glycosylation-targeted therapies are discussed. By integrating these findings, this review aims to provide a comprehensive perspective to further understand the role of aberrant glycosylation modifications in the pathophysiology of SCZ.
Collapse
Affiliation(s)
- Yanchen Feng
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou, China
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lu Sun
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xue Dang
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Diyan Liu
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ziyun Liao
- College of Acupuncture, Moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jianping Yao
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yunke Zhang
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ziqi Deng
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinyao Li
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Min Zhao
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou, China
- Hospital of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Feixiang Liu
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou, China
- Hospital of Encephalopathy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
17
|
Li J, Wang Y, Yang Y, Ren X, Qiang Y, Zhang L, Guo L, Liu K. Reactive astrogliosis induced by TNF-α is associated with upregulated AEG-1 together with activated NF-κB pathway in vitro. Neurosci Lett 2024; 837:137899. [PMID: 39019146 DOI: 10.1016/j.neulet.2024.137899] [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: 04/28/2024] [Revised: 07/09/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
Astrocyte-elevated gene-1 (AEG-1/MTDH/LYRIC) has garnered signficant attention in cancer research, yet, its role in inflammation-associated astrogliosis remains underexplored. This study aims to elucidate the effects of AEG-1 on reactive astrogliosis, including proliferation, migration, and glutamate uptake in primary astrocytes derived from rats. We first confirmed the effect of AEG-1 on these parameters. Subsequently, we investigated whether AEG-1 plays a role in the process of pro-inflammation factors such as tumor necrosis factor-alpha (TNF-α) induced astrogliosis. Our findings revealed that AEG-1-lentivirus infection led to hypertrophic cell bodies and enhanced expression of astrogliosis markers, including glial fibrillary acidic protein (GFAP) and vimentin. Additionally, AEG-1 was found to upregulate the mRNA and protein expression levels of EAAT2, a major glutamate transporter in the brain predominantly expressed by astrocytes and responsible for 90% of glutamate clearance. Furthermore, TNF-α was shown to promote astrogliosis, as well as astrocyte proliferation and migration, by upregulating AEG-1 expression through the NF-κB pathway. Collectively, these results suggest a potential role for AEG-1 in inflammation-related astrogliosis.
Collapse
Affiliation(s)
- Juanjuan Li
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan 750004, China.
| | - Yahe Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan 750004, China.
| | - Yong Yang
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan 750004, China.
| | - Xiaofan Ren
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan 750004, China.
| | - Yuanyuan Qiang
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan 750004, China.
| | - Lianxiang Zhang
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan 750004, China.
| | - Le Guo
- School of Laboratory Medicine, Ningxia Medical University, Yinchuan 750004, China.
| | - Kunmei Liu
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
18
|
Poljak L, Miše B, Čičin-Šain L, Tvrdeić A. Ceftriaxone Inhibits Conditioned Fear and Compulsive-like Repetitive Marble Digging without Central Nervous System Side Effects Typical of Diazepam-A Study on DBA2/J Mice and a High-5HT Subline of Wistar-Zagreb 5HT Rats. Biomedicines 2024; 12:1711. [PMID: 39200176 PMCID: PMC11351474 DOI: 10.3390/biomedicines12081711] [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: 05/16/2024] [Revised: 07/14/2024] [Accepted: 07/22/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Ceftriaxone upregulates GLT1 glutamate transporter in the brain and may have anti-CFC and anti-OCD effects. Methods: Twenty WZ-5HT rats were used to investigate the effects of ceftriaxone on obsessive-compulsive (OCD)-like behaviour in the marble-burying (MB) test, freezing behaviour in contextual fear conditioning (CFC) and expression of GLT1 protein in the hippocampus or amygdala using immunoblots. Fifteen DBA/2J mice were used in the MB test. We also compared diazepam with ceftriaxone in open-field, beam-walking, and wire-hanging tests on 47 DBA/2J mice. Ceftriaxone (200 mg/kg) and saline were applied intraperitoneally, once daily for 7 (rats) or 5 (mice) consecutive days. A single dose of diazepam (1.5-3.0 mg/kg) or saline was injected 30 min before the behavioural tests. Results: Ceftriaxone significantly diminished OCD-like behaviour (↓ number of marbles buried) and freezing behaviour in CFC context session (↑ latencies, ↓ total duration, ↓ duration over four 2 min periods of the session) but increased GLT1 protein expression in the amygdala and hippocampus of rats. Diazepam induced sedation, ataxia and myorelaxation in mice. Ceftriaxone did not have these side effects. Conclusions: The results of this study confirm the anti-CFC and anti-OCD effects of ceftriaxone, which did not produce the unwanted effects typical of diazepam.
Collapse
Affiliation(s)
- Ljiljana Poljak
- Department of Physiology and Immunology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Branko Miše
- University Hospital for Infectious Diseases “Fran Mihaljević”, 10000 Zagreb, Croatia;
| | - Lipa Čičin-Šain
- Laboratory for Neurochemistry and Molecular Neurobiology, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Ante Tvrdeić
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
19
|
Ramazi S, Dadzadi M, Darvazi M, Seddigh N, Allahverdi A. Protein modification in neurodegenerative diseases. MedComm (Beijing) 2024; 5:e674. [PMID: 39105197 PMCID: PMC11298556 DOI: 10.1002/mco2.674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Posttranslational modifications play a crucial role in governing cellular functions and protein behavior. Researchers have implicated dysregulated posttranslational modifications in protein misfolding, which results in cytotoxicity, particularly in neurodegenerative diseases such as Alzheimer disease, Parkinson disease, and Huntington disease. These aberrant posttranslational modifications cause proteins to gather in certain parts of the brain that are linked to the development of the diseases. This leads to neuronal dysfunction and the start of neurodegenerative disease symptoms. Cognitive decline and neurological impairments commonly manifest in neurodegenerative disease patients, underscoring the urgency of comprehending the posttranslational modifications' impact on protein function for targeted therapeutic interventions. This review elucidates the critical link between neurodegenerative diseases and specific posttranslational modifications, focusing on Tau, APP, α-synuclein, Huntingtin protein, Parkin, DJ-1, and Drp1. By delineating the prominent aberrant posttranslational modifications within Alzheimer disease, Parkinson disease, and Huntington disease, the review underscores the significance of understanding the interplay among these modifications. Emphasizing 10 key abnormal posttranslational modifications, this study aims to provide a comprehensive framework for investigating neurodegenerative diseases holistically. The insights presented herein shed light on potential therapeutic avenues aimed at modulating posttranslational modifications to mitigate protein aggregation and retard neurodegenerative disease progression.
Collapse
Affiliation(s)
- Shahin Ramazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Maedeh Dadzadi
- Department of BiotechnologyFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mona Darvazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Nasrin Seddigh
- Department of BiochemistryFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Abdollah Allahverdi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| |
Collapse
|
20
|
Liu L, Tian X, Li W. Mechanistic study of the anti-excitatory amino acid toxicity of Bushen Zhichan decoction for Parkinson's disease based on the transcriptional regulation of EAAT1 by YY1. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117857. [PMID: 38350506 DOI: 10.1016/j.jep.2024.117857] [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: 11/08/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bushen Zhichan decoction (BSZCF) is derived from Liuwei Dihuang Pill, a famous Chinese herbal formula recorded in the book Key to Therapeutics of Children's Diseases. It has been widely used as a basic prescription for nourishing and tonifying the liver and kidneys to treat Parkinson's disease (PD), but its mechanism remains to be explored. AIM OF THE STUDY BSZCF, a Chinese herbal formula comprising five herbs: Rehmannia glutinosa (Gaertn.) DC., Dioscorea oppositifolia L., Cornus officinalis Siebold & Zucc., Fallopia multiflora (Thunb.) Haraldson and Cistanche tubulosa (Schenk) Wight, is used clinically to treat PD. In vivo and in vitro experiments were designed to elucidate the mechanism of BSZCF in the protection of dopamine (DA) neurons and the treatment of PD. The toxicity of excitatory amino acids (EAA) may be attenuated by inhibiting the transcription factor Yin Yang 1 (YY1) and up-regulating the expression of excitatory amino acid transporter 1 (EAAT1). MATERIALS AND METHODS IN VIVO: After 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was intraperitoneally injected into specific pathogen free (SPF) C57BL/6J mice, model mice were intragastrically given adamantane hydrochloride tablets (AHT) or different doses of BSZCF for 14 days. Both open field and pole-climbing tests were conducted to assess behavioral changes. In vitro: 1-Methyl-4-phe-nylpyridiniumiodide (MPP+)-injured human neuroblastoma cells (SH-SY5Y) were utilized to construct PD cell models. Primary astrocytes were transfected with EAAT1 and YY1 lentiviruses for EAAT1 gene knockout and YY1 gene knockout astrocytes, respectively. The high performance liquid chromatography-mass spectrometry (HPLC-MS) analysis of BSZCF was performed to control the quality of blood drugs. The optimal concentration and time of PD cell models treated by BSZCF were determined by the use of Cell Counting Kit-8 (CCK8). Enzyme-linked immunosorbent assay (ELISA) was used for measuring glutamate (Glu) in the peripheral blood and cells of each group. Western blotting (WB) and real-time quantitative polymerase chain reaction (qPCR) were used to detect tyrosine hydroxylase (TH), dopamine transporters (DAT), EAAT1 and YY1 protein and mRNA. After the blockade of EAAT1, immunofluorescence (IF) assay was used to detect the TH protein in each group. RESULTS In vivo research showed that BSZCF improved the behavioral symptoms of PD mice, and reduced the death of DA neurons and the level of Glu. The mechanism may be related to the decrease of YY1 expression and the increase of EAAT1 levels. In vitro experiments showed that the anti-excitatory amino acid toxicity of BSZCF was achieved by inhibiting YY1 expression and regulating EAAT1. CONCLUSIONS By inhibiting YY1 to increase the expression of EAAT1 and attenuating the toxicity of Glu, BSZCF exerts the effect of protecting DA neurons and treating PD-like symptoms in mice.
Collapse
Affiliation(s)
- Leilei Liu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| | - Xinyun Tian
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| | - Wentao Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| |
Collapse
|
21
|
Firth W, Pye KR, Weightman Potter PG. Astrocytes at the intersection of ageing, obesity, and neurodegeneration. Clin Sci (Lond) 2024; 138:515-536. [PMID: 38652065 DOI: 10.1042/cs20230148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Once considered passive cells of the central nervous system (CNS), glia are now known to actively maintain the CNS parenchyma; in recent years, the evidence for glial functions in CNS physiology and pathophysiology has only grown. Astrocytes, a heterogeneous group of glial cells, play key roles in regulating the metabolic and inflammatory landscape of the CNS and have emerged as potential therapeutic targets for a variety of disorders. This review will outline astrocyte functions in the CNS in healthy ageing, obesity, and neurodegeneration, with a focus on the inflammatory responses and mitochondrial function, and will address therapeutic outlooks.
Collapse
Affiliation(s)
- Wyn Firth
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, U.K
| | - Katherine R Pye
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Paul G Weightman Potter
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| |
Collapse
|
22
|
Karagöl A, Karagöl T, Smorodina E, Zhang S. Structural bioinformatics studies of glutamate transporters and their AlphaFold2 predicted water-soluble QTY variants and uncovering the natural mutations of L->Q, I->T, F->Y and Q->L, T->I and Y->F. PLoS One 2024; 19:e0289644. [PMID: 38598436 PMCID: PMC11006163 DOI: 10.1371/journal.pone.0289644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/22/2023] [Indexed: 04/12/2024] Open
Abstract
Glutamate transporters play key roles in nervous physiology by modulating excitatory neurotransmitter levels, when malfunctioning, involving in a wide range of neurological and physiological disorders. However, integral transmembrane proteins including the glutamate transporters remain notoriously difficult to study, due to their localization within the cell membrane. Here we present the structural bioinformatics studies of glutamate transporters and their water-soluble variants generated through QTY-code, a protein design strategy based on systematic amino acid substitutions. These include 2 structures determined by X-ray crystallography, cryo-EM, and 6 predicted by AlphaFold2, and their predicted water-soluble QTY variants. In the native structures of glutamate transporters, transmembrane helices contain hydrophobic amino acids such as leucine (L), isoleucine (I), and phenylalanine (F). To design water-soluble variants, these hydrophobic amino acids are systematically replaced by hydrophilic amino acids, namely glutamine (Q), threonine (T) and tyrosine (Y). The QTY variants exhibited water-solubility, with four having identical isoelectric focusing points (pI) and the other four having very similar pI. We present the superposed structures of the native glutamate transporters and their water-soluble QTY variants. The superposed structures displayed remarkable similarity with RMSD 0.528Å-2.456Å, despite significant protein transmembrane sequence differences (41.1%->53.8%). Additionally, we examined the differences of hydrophobicity patches between the native glutamate transporters and their QTY variants. Upon closer inspection, we discovered multiple natural variations of L->Q, I->T, F->Y and Q->L, T->I, Y->F in these transporters. Some of these natural variations were benign and the remaining were reported in specific neurological disorders. We further investigated the characteristics of hydrophobic to hydrophilic substitutions in glutamate transporters, utilizing variant analysis and evolutionary profiling. Our structural bioinformatics studies not only provided insight into the differences between the hydrophobic helices and hydrophilic helices in the glutamate transporters, but they are also expected to stimulate further study of other water-soluble transmembrane proteins.
Collapse
Affiliation(s)
- Alper Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Taner Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Eva Smorodina
- Laboratory for Computational and Systems Immunology, Department of Immunology, University of Oslo, Oslo University Hospital, Oslo, Norway
| | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| |
Collapse
|
23
|
Long J, Song X, Wang C, Peng L, Niu L, Li Q, Huang R, Zhang R. Global-brain functional connectivity related with trait anxiety and its association with neurotransmitters and gene expression profiles. J Affect Disord 2024; 348:248-258. [PMID: 38159654 DOI: 10.1016/j.jad.2023.12.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/30/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Numerous studies have explored the neural correlates of trait anxiety, a predisposing factor for several stress-related disorders. However, the findings from previous studies are inconsistent, which might be due to the limited regions of interest (ROI). A recent approach, named global-brain functional connectivity (GBC), has been demonstrated to address the shortcomings of ROI-based analysis. Furthermore, research on the transcriptome-connectome association has provided an approach to link the microlevel transcriptome profile with the macroscale brain network. In this paper, we aim to explore the neurobiology of trait anxiety with an imaging transcriptomic approach using GBC, biological neurotransmitters, and transcriptome profiles. METHODS Using a sample of resting-state fMRI data, we investigated trait anxiety-related alteration in GBC. We further used behavioral analysis, spatial correlation analysis, and postmortem gene expression to separately assess the cognitive functions, neurotransmitters, and transcriptional profiles related to alteration in GBC in individuals with trait anxiety. RESULTS GBC values in the ventromedial prefrontal cortex and the precuneus were negatively correlated with levels of trait anxiety. This alteration was correlated with behavioral terms including social cognition, emotion, and memory. A strong association was revealed between trait anxiety-related alteration in GBC and neurotransmitters, including dopaminergic, serotonergic, GABAergic, and glutamatergic systems in the ventromedial prefrontal cortex and the precuneus. The transcriptional profiles explained the functional connectivity, with correlated genes enriched in transmembrane signaling. LIMITATIONS Several limitations should be taken into account in this research. For example, future research should consider using some different approaches based on dynamic or task-based functional connectivity analysis, include more neurotransmitter receptors, additional gene expression data from different samples or more genes related to other stress-related disorders. Meanwhile, it is of great significance to include a larger sample size of individuals with a diagnosis of major depression disorder or other disorders for analysis and comparison and apply stricter multiple-comparison correction and threshold settings in future research. CONCLUSIONS Our research employed multimodal data to investigate GBC in the context of trait anxiety and to establish its associations with neurotransmitters and transcriptome profiles. This approach may improve understanding of the neural mechanism, together with the biological and molecular genetic foundations of GBC in trait anxiety.
Collapse
Affiliation(s)
- Jixin Long
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoqi Song
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Chanyu Wang
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China; Faculty of Medicine and Health Sciences, Department of Head and Skin, Ghent Experimental Psychiatry (GHEP) lab, Ghent University, Ghent, Belgium
| | - Lanxin Peng
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lijing Niu
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qian Li
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Ruiwang Huang
- School of Psychology, South China Normal University, Guangzhou, China
| | - Ruibin Zhang
- Laboratory of Cognitive Control and Brain Healthy, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China; Department of Psychiatry, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
24
|
Vazquez-Juarez E, Srivastava I, Lindskog M. The effect of ketamine on synaptic mistuning induced by impaired glutamate reuptake. Neuropsychopharmacology 2023; 48:1859-1868. [PMID: 37301901 PMCID: PMC10584870 DOI: 10.1038/s41386-023-01617-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/05/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023]
Abstract
Mistuning of synaptic transmission has been proposed to underlie many psychiatric disorders, with decreased reuptake of the excitatory neurotransmitter glutamate as one contributing factor. Synaptic tuning occurs through several diverging and converging forms of plasticity. By recording evoked field postsynaptic potentials in the CA1 area in hippocampal slices, we found that inhibiting glutamate transporters using DL-TBOA causes retuning of synaptic transmission, resulting in a new steady state with reduced synaptic strength and a lower threshold for inducing long-term synaptic potentiation (LTP). Moreover, a similar reduced threshold for LTP was observed in a rat model of depression with decreased levels of glutamate transporters. Most importantly, we found that the antidepressant ketamine counteracts the effects of increased glutamate on the various steps involved in synaptic retuning. We, therefore, propose that ketamine's mechanism of action as an antidepressant is to restore adequate synaptic tuning.
Collapse
Affiliation(s)
- Erika Vazquez-Juarez
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ipsit Srivastava
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden
- Department of Medical Cell Biology, Uppsala University, 751 24, Uppsala, Sweden
| | - Maria Lindskog
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77, Stockholm, Sweden.
- Department of Medical Cell Biology, Uppsala University, 751 24, Uppsala, Sweden.
| |
Collapse
|
25
|
Pańczyszyn-Trzewik P, Czechowska E, Stachowicz K, Sowa-Kućma M. The Importance of α-Klotho in Depression and Cognitive Impairment and Its Connection to Glutamate Neurotransmission-An Up-to-Date Review. Int J Mol Sci 2023; 24:15268. [PMID: 37894946 PMCID: PMC10607524 DOI: 10.3390/ijms242015268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/11/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
Depression is a serious neuropsychiatric disease affecting an increasing number of people worldwide. Cognitive deficits (including inattention, poor memory, and decision-making difficulties) are common in the clinical picture of depression. Cognitive impairment has been hypothesized to be one of the most important components of major depressive disorder (MDD; referred to as clinical depression), although typical cognitive symptoms are less frequent in people with depression than in people with schizophrenia or bipolar disorder (BD; sometimes referred to as manic-depressive disorder). The importance of α-Klotho in the aging process has been well-documented. Growing evidence points to the role of α-Klotho in regulating other biological functions, including responses to oxidative stress and the modulation of synaptic plasticity. It has been proven that a Klotho deficit may contribute to the development of various nervous system pathologies, such as behavioral disorders or neurodegeneration. Given the growing evidence of the role of α-Klotho in depression and cognitive impairment, it is assumed that this protein may be a molecular link between them. Here, we provide a research review of the role of α-Klotho in depression and cognitive impairment. Furthermore, we propose potential mechanisms (related to oxidative stress and glutamatergic transmission) that may be important in α-Klotho-mediated regulation of mental and cognitive function.
Collapse
Affiliation(s)
- Patrycja Pańczyszyn-Trzewik
- Department of Human Physiology, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland; (P.P.-T.); (E.C.)
| | - Ewelina Czechowska
- Department of Human Physiology, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland; (P.P.-T.); (E.C.)
| | - Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland;
| | - Magdalena Sowa-Kućma
- Department of Human Physiology, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland; (P.P.-T.); (E.C.)
- Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, Warzywna Street 1A, 35-595 Rzeszow, Poland
| |
Collapse
|
26
|
Costa J, Hayes C, Lisacek F. Protein glycosylation and glycoinformatics for novel biomarker discovery in neurodegenerative diseases. Ageing Res Rev 2023; 89:101991. [PMID: 37348818 DOI: 10.1016/j.arr.2023.101991] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/25/2023] [Accepted: 06/18/2023] [Indexed: 06/24/2023]
Abstract
Glycosylation is a common post-translational modification of brain proteins including cell surface adhesion molecules, synaptic proteins, receptors and channels, as well as intracellular proteins, with implications in brain development and functions. Using advanced state-of-the-art glycomics and glycoproteomics technologies in conjunction with glycoinformatics resources, characteristic glycosylation profiles in brain tissues are increasingly reported in the literature and growing evidence shows deregulation of glycosylation in central nervous system disorders, including aging associated neurodegenerative diseases. Glycan signatures characteristic of brain tissue are also frequently described in cerebrospinal fluid due to its enrichment in brain-derived molecules. A detailed structural analysis of brain and cerebrospinal fluid glycans collected in publications in healthy and neurodegenerative conditions was undertaken and data was compiled to create a browsable dedicated set in the GlyConnect database of glycoproteins (https://glyconnect.expasy.org/brain). The shared molecular composition of cerebrospinal fluid with brain enhances the likelihood of novel glycobiomarker discovery for neurodegeneration, which may aid in unveiling disease mechanisms, therefore, providing with novel therapeutic targets as well as diagnostic and progression monitoring tools.
Collapse
Affiliation(s)
- Júlia Costa
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal.
| | - Catherine Hayes
- Proteome Informatics Group, Swiss Institute of Bioinformatics, CH-1227 Geneva, Switzerland
| | - Frédérique Lisacek
- Proteome Informatics Group, Swiss Institute of Bioinformatics, CH-1227 Geneva, Switzerland; Computer Science Department, University of Geneva, CH-1227 Geneva, Switzerland; Section of Biology, University of Geneva, CH-1211 Geneva, Switzerland
| |
Collapse
|
27
|
Tarkany Basting R, Henrique Napimoga M, Antônio Trindade Silva C, Ballassini Abdalla H, Campos Durso B, Henrique Barboza Martins L, de Abreu Cavalcanti H, Hammock BD, Trindade Clemente-Napimoga J. Soluble epoxide hydrolase inhibitor blockage microglial cell activation in subnucleus caudalis in a persistent model of arthritis. Int Immunopharmacol 2023; 120:110320. [PMID: 37230034 PMCID: PMC10631565 DOI: 10.1016/j.intimp.2023.110320] [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: 02/14/2023] [Revised: 04/30/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic condition characterized by pain and infiltration of immune cells into the joint. Immune cells can be activated, producing inflammatory cytokines, leading to continuously degenerative and inflammatory reactions and the temporomandibular joint (TMJ) can be affected by RA. In this scenario, novel targets are needed to increase treatment efficacy with minimized side effects. The epoxy-eicosatrienoic acids (EETs), are endogenous signaling molecules, playing important roles in diminishing inflammation and pain but are promptly metabolized by soluble epoxide hydrolase (sEH), generating less-bioactive acids.Therefore, sEH inhibitors is an interest therapeutic target to enhance the beneficial effect of natural EETs. TPPU is a potent sEH inhibitor that is capable of dampening EETs hydrolysis. Thus, we aimed to assess the impact of pharmacological sEH inhibition on a persistent model of albumin-induced arthritis in the TMJ, in two scenarios: first, as post-treatment, in an installed arthritic condition, and second, the protective role, in preventing the development of an arthritic condition. In addition, we investigate the influence of sEH inhibition on microglia cell activation in the trigeminal subnucleus caudalis (TSC) and in vitro experiments. Finally, we examined the astrocyte phenotype. Oral administration of TPPU, acts in multiple pathways, in a protective and reparative post-treatment, ameliorating the preservation of the TMJ morphology, reducing the hypernociception, with an immunosuppressive action reducing neutrophil and lymphocytes and pro-inflammatory cytokines in the TMJ of rats. In TSC, TPPU reduces the cytokine storm and attenuates the microglia activated P2X7/Cathepsin S/Fractalkine pathway and reduces the astrocyte activation and glutamate levels. Collectively, our findings revealed that sEH inhibition mitigates hypersensitive nociception through the regulation of microglia activation and astrocyte modulation, demonstrating the potential use of sEH inhibitors as immunoresolvents in the treatment of autoimmune disorders.
Collapse
Affiliation(s)
- Rosanna Tarkany Basting
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Marcelo Henrique Napimoga
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Carlos Antônio Trindade Silva
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Henrique Ballassini Abdalla
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Braz Campos Durso
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | | | - Herbert de Abreu Cavalcanti
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA, United States of America; EicOsis LLC, Davis, CA, United States of America
| | | |
Collapse
|
28
|
Kamalmaz N, Ben Bacha A, Alonazi M, Albasher G, Khayyat AIA, El-Ansary A. Unveiling sex-based differences in developing propionic acid-induced features in mice as a rodent model of ASD. PeerJ 2023; 11:e15488. [PMID: 37334116 PMCID: PMC10274690 DOI: 10.7717/peerj.15488] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Background Males are more likely to develop autism as a neurodevelopmental disorder than females are, although the mechanisms underlying male vulnerability are not fully understood. Therefore, studying the role of autism etiologies considering sex differences in the propionic acid (PPA) rodent model of autism would build greater understanding of how females are protected from autism spectrum disorder, which may be used as a treatment strategy for males with autism. Objectives This study aimed to investigate the sex differences in oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut microbiota impairment as etiological mechanisms for many neurological diseases, with specific reference to autism. Method Forty albino mice were divided into four groups of 10 animals each with two control and two treated groups of both sexes received only phosphate-buffered saline or a neurotoxic dose of PPA (250 mg/kg body weight) for 3 days, respectively. Biochemical markers of energy metabolism, oxidative stress, neuroinflammation, and excitotoxicity were measured in mouse brain homogenates, whereas the presence of pathogenic bacteria was assessed in mouse stool samples. Furthermore, the repetitive behavior, cognitive ability, and physical-neural coordination of the animals were examined. Results Collectively, selected variables related to oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut bacteria were impaired concomitantly with altered behavior in PPA-induced rodent model, with males being more susceptible than females. Conclusion This study explains the role of sex in the higher vulnerability of males to develop autistic biochemical and behavioral features compared with females. Female sex hormones and the higher detoxification capacity and higher glycolytic flux in females serve as neuroprotective contributors in a rodent model of autism.
Collapse
Affiliation(s)
- Nasreen Kamalmaz
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Gadah Albasher
- Zoology Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Arwa Ishaq A. Khayyat
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
29
|
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
|
30
|
Colovic M, Yang H, Southcott L, Merkens H, Colpo N, Bénard F, Schaffer P. Comparative Evaluation of [ 18F]5-Fluoroaminosuberic Acid and (4 S)-4-3-[ 18F]fluoropropyl)-l-Glutamate as System xC--Targeting Radiopharmaceuticals. J Nucl Med 2023:jnumed.122.265254. [PMID: 37116917 DOI: 10.2967/jnumed.122.265254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/21/2023] [Indexed: 04/30/2023] Open
Abstract
System [Formula: see text] is an appealing biomarker for targeting oxidative stress with oncologic PET imaging and can serve as an alternative PET biomarker to other metabolic indicators. In this paper, we report a direct comparison of 2 18F-labeled amino acid radiopharmaceuticals targeting system [Formula: see text], [18F]5-fluoroaminosuberic acid ([18F]FASu) and (4S)-4-(3-[18F]fluoropropyl)-l-glutamate ([18F]FSPG), in terms of their uptake specificity and ability to image glioma and lung cancer xenografts in vivo. Methods: Both tracers were synthesized according to previously published procedures. In vitro uptake specificity assays were conducted using prostate (PC-3), glioblastoma (U-87), colorectal (HT-29), ovarian (SKOV3), breast (MDA-MB-231), and lung cancer (A549) cell lines. PET/CT imaging and biodistribution studies were conducted in immunocompromised mice bearing U-87 or A549 xenografts. Results: In vitro cell uptake assays showed that the tracers accumulated in cancer cells in a time-dependent manner and that the uptake of [18F]FASu was blocked by the system [Formula: see text] inhibitor sulfasalazine and rose bengal, but not by system L inhibitor 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid, system [Formula: see text] inhibitor L-trans-pyrrolidine-2,4-dicarboxylic acid, or l-serine, which is a substrate for transporter systems A, ACS, B0, and B0,+ Conversely, [18F]FSPG uptake decreased significantly in the presence of an excess of L-trans-pyrrolidine-2,4-dicarboxylic acid in 2 of 3 tested cell lines, indicating some reliance on system [Formula: see text] in these cells. In an in vivo setting, [18F]FASu and [18F]FSPG generated good-contrast PET images in U-87 and A549 tumor-bearing mice. Tracer accumulation in A549 tumors was 5.0 ± 0.8 percentage injected dose (%ID)/g ([18F]FASu, n ≥ 5) and 6.3 ± 1.3 %ID/g ([18F]FSPG, n ≥ 6, P = 0.7786), whereas U-87 xenografts demonstrated uptake of 6.1 ± 2.4 %ID/g ([18F]FASu, n ≥ 4) and 11.2 ± 4.1 %ID/g ([18F]FSPG, n ≥ 4, P = 0.0321) at 1 h after injection. Conclusion: [18F]FSPG had greater in vitro uptake than [18F]FASu in all cell lines tested; however, our results indicate that residual uptake differences exist between [18F]FSPG and [18F]FASu, suggesting alternative transporter activity in the cell lines tested. In vivo studies demonstrated the ability of both [18F]FASu and [18F]FSPG to image glioblastoma (U-87) and non-small cell lung cancer (A549) xenografts.
Collapse
Affiliation(s)
- Milena Colovic
- Life Sciences Division, TRIUMF, Vancouver, British Columbia, Canada
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF, Vancouver, British Columbia, Canada
| | - Lily Southcott
- Life Sciences Division, TRIUMF, Vancouver, British Columbia, Canada
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Nadine Colpo
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Francois Bénard
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, Vancouver, British Columbia, Canada;
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada; and
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
31
|
Nguyen A, Patel AB, Kioutchoukova IP, Diaz MJ, Lucke-Wold B. Mechanisms of Mitochondrial Oxidative Stress in Brain Injury: From Pathophysiology to Therapeutics. OXYGEN 2023; 3:163-178. [PMID: 37082315 PMCID: PMC10111246 DOI: 10.3390/oxygen3020012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Mitochondrial oxidative stress has been implicated in various forms of brain injury, both traumatic and non-traumatic. Due to its oxidative demand, the brain is intimately dependent on its mitochondrial functioning. However, there remains appreciable heterogeneity in the development of these injuries regarding ROS and their effect on the sequelae. These include traumatic insults such as TBIs and intracranial hemorrhaging secondary to this. In a different vein, such injuries may be attributed to other etiologies such as infection, neoplasm, or spontaneous hemorrhage (strokes, aneurysms). Clinically, the manner of treatment may also be adjusted in relation to each injury and its unique progression in the context of ROS. In the current review, then, the authors highlight the role of mitochondrial ROS in various forms of brain injury, emphasizing both the collective and unique elements of each form. Lastly, these narratives are met with the current therapeutic landscape and the role of emerging therapies in treating reactive oxygen species in brain injuries.
Collapse
Affiliation(s)
- Andrew Nguyen
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Anjali B. Patel
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | - Michael J. Diaz
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, 1600 SW Archer Rd., Gainesville, FL 32610, USA
| |
Collapse
|
32
|
Gao C, Yang B, Li Y, Pei W. Monocarboxylate transporter-dependent mechanism is involved in the adaptability of the body to exercise-induced fatigue under high-altitude hypoxia environment. Brain Res Bull 2023; 195:78-85. [PMID: 36804772 DOI: 10.1016/j.brainresbull.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/28/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
Under high-altitude hypoxia environment, the body is more prone to fatigue, which occurs in both peripheral muscles and the central nervous system (CNS). The key factor determining the latter is the imbalance of brain energy metabolism, which makes it difficult to maintain the central nervous system to send peripheral nerve impulse continuously. During strenuous exercise, lactate released from astrocytes is taken up by neurons stored for energy to maintain synaptic transmission, a process mediated by monocarboxylate transporters (MCTs) in CNS. The present study investigated the correlation among the adaptability to exercise-induced fatigue, brain lactate metabolism and neuronal hypoxia injury under high-altitude hypoxia environment. Rats were subjected to exhaustive incremental load treadmill exercise under either normal pressure and normoxic conditions or simulated high-altitude low pressure and hypoxic conditions, with subsequent evaluation of the average exhaustive time as well as the expression of monocarboxylate transporters 2 (MCT2), MCT4, the average neuronal density in the cerebral motor cortex, and the lactate content in rat brain. At the early stage of simulated high-altitude environment, the average exhaustive time and neuronal density of rats decreased rapidly, then gradually recovered to some extent with the extension of altitude acclimatization time. The expression of MCT2, MCT4 and the lactate content in rat brain also increased gradually with the extension of altitude acclimatization time. After the application of lactate transport inhibitor, the recovery of exercise capacity of rats after altitude acclimatization was quickly blocked, and the neuronal injury in the cerebral motor cortex of rats was also significantly aggravated. These findings demonstrate that MCT-dependent mechanism is involved in the adaptability of the body to central fatigue, and provide a potential basis for medical intervention for exercise-induced fatigue under high-altitude hypoxia environment.
Collapse
Affiliation(s)
- Chen Gao
- Department of General Practice, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, China.
| | - Binni Yang
- Department of General Practice, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, China
| | - Yurong Li
- Department of General Practice, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, China
| | - Wenjuan Pei
- Department of General Practice, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, China
| |
Collapse
|
33
|
Tang XH, Diao YG, Ren ZY, Zang YY, Zhang GF, Wang XM, Duan GF, Shen JC, Hashimoto K, Zhou ZQ, Yang JJ. A role of GABA A receptor α1 subunit in the hippocampus for rapid-acting antidepressant-like effects of ketamine. Neuropharmacology 2023; 225:109383. [PMID: 36565851 DOI: 10.1016/j.neuropharm.2022.109383] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Ketamine can produce rapid-acting antidepressant effects in treatment-resistant patients with depression. Although alterations in glutamatergic and GABAergic neurotransmission in the brain play a role in depression, the precise molecular mechanisms in these neurotransmission underlying ketamine's antidepressant actions remain largely unknown. Mice exposed to FSS (forced swimming stress) showed depression-like behavior and decreased levels of GABA (γ-aminobutyric acid), but not glutamate, in the hippocampus. Ketamine increased GABA levels and decreased glutamate levels in the hippocampus of mice exposed to FSS. There was a correlation between GABA levels and depression-like behavior. Furthermore, ketamine increased the levels of enzymes and transporters on the GABAergic neurons (SAT1, GAD67, GAD65, VGAT and GAT1) and astrocytes (EAAT2 and GAT3), without affecting the levels of enzymes and transporters (SAT2, VGluT1 and GABAAR γ2) on glutamatergic neurons. Moreover, ketamine caused a decreased expression of GABAAR α1 subunit, which was specifically expressed on GABAergic neurons and astrocytes, an increased GABA synthesis and metabolism in GABAergic neurons, a plasticity change in astrocytes, and an increase in ATP (adenosine triphosphate) contents. Finally, GABAAR antagonist bicuculline or ATP exerted a rapid antidepressant-like effect whereas pretreatment with GABAAR agonist muscimol blocked the antidepressant-like effects of ketamine. In addition, pharmacological activation and inhibition of GABAAR modulated the synthesis and metabolism of GABA, and the plasticity of astrocytes in the hippocampus. The present data suggest that ketamine could increase GABA synthesis and astrocyte plasticity through downregulation of GABAAR α1, increases in GABA, and conversion of GABA into ATP, resulting in a rapid-acting antidepressant-like action. This article is part of the Special Issue on 'Ketamine and its Metabolites'.
Collapse
Affiliation(s)
- Xiao-Hui Tang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu-Gang Diao
- Department of Anesthesiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Zhuo-Yu Ren
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan-Yu Zang
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Guang-Fen Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xing-Ming Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Gui-Fang Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Jin-Chun Shen
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Zhi-Qiang Zhou
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
34
|
Chakraborty P, Dey A, Gopalakrishnan AV, Swati K, Ojha S, Prakash A, Kumar D, Ambasta RK, Jha NK, Jha SK, Dewanjee S. Glutamatergic neurotransmission: A potential pharmacotherapeutic target for the treatment of cognitive disorders. Ageing Res Rev 2023; 85:101838. [PMID: 36610558 DOI: 10.1016/j.arr.2022.101838] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023]
Abstract
In the mammalian brain, glutamate is regarded to be the primary excitatory neurotransmitter due to its widespread distribution and wide range of metabolic functions. Glutamate plays key roles in regulating neurogenesis, synaptogenesis, neurite outgrowth, and neuron survival in the brain. Ionotropic and metabotropic glutamate receptors, neurotransmitters, neurotensin, neurosteroids, and others co-ordinately formulate a complex glutamatergic network in the brain that maintains optimal excitatory neurotransmission. Cognitive activities are potentially synchronized by the glutamatergic activities in the brain via restoring synaptic plasticity. Dysfunctional glutamate receptors and other glutamatergic components are responsible for the aberrant glutamatergic activity in the brain that cause cognitive impairments, loss of synaptic plasticity, and neuronal damage. Thus, controlling the brain's glutamatergic transmission and modifying glutamate receptor function could be a potential therapeutic strategy for cognitive disorders. Certain drugs that regulate glutamate receptor activities have shown therapeutic promise in improving cognitive functions in preclinical and clinical studies. However, several issues regarding precise functional information of glutamatergic activity are yet to be comprehensively understood. The present article discusses the scope of developing glutamatergic systems as prospective pharmacotherapeutic targets to treat cognitive disorders. Special attention has been given to recent developments, challenges, and future prospects.
Collapse
Affiliation(s)
- Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, West Bengal, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Kumari Swati
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Anand Prakash
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand 248007, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India.
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
35
|
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
|
36
|
Gao C, Yang B, Li Y, Pei W. A monocarboxylate transporter-dependent mechanism confers resistance to exercise-induced fatigue in a high-altitude hypoxic environment. Sci Rep 2023; 13:2949. [PMID: 36807596 PMCID: PMC9941081 DOI: 10.1038/s41598-023-30093-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The body is more prone to fatigue in a high-altitude hypoxic environment, in which fatigue occurs in both peripheral muscles and the central nervous system (CNS). The key factor determining the latter is the imbalance in brain energy metabolism. During strenuous exercise, lactate released from astrocytes is taken up by neurons via monocarboxylate transporters (MCTs) as a substrate for energy metabolism. The present study investigated the correlations among the adaptability to exercise-induced fatigue, brain lactate metabolism and neuronal hypoxia injury in a high-altitude hypoxic environment. Rats were subjected to exhaustive incremental load treadmill exercise under either normal pressure and normoxic conditions or simulated high-altitude, low-pressure and hypoxic conditions, with subsequent evaluation of the average exhaustive time as well as the expression of MCT2 and MCT4 in the cerebral motor cortex, the average neuronal density in the hippocampus, and the brain lactate content. The results illustrate that the average exhaustive time, neuronal density, MCT expression and brain lactate content were positively correlated with the altitude acclimatization time. These findings demonstrate that an MCT-dependent mechanism is involved in the adaptability of the body to central fatigue and provide a potential basis for medical intervention for exercise-induced fatigue in a high-altitude hypoxic environment.
Collapse
Affiliation(s)
- Chen Gao
- Department of General Practice, The 940Th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, BinHe South Road, No.333, Lanzhou, 730050, Gansu, China.
| | - Binni Yang
- Department of General Practice, The 940Th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, BinHe South Road, No.333, Lanzhou, 730050 Gansu China
| | - Yurong Li
- Department of General Practice, The 940Th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, BinHe South Road, No.333, Lanzhou, 730050 Gansu China
| | - Wenjuan Pei
- Department of General Practice, The 940Th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, BinHe South Road, No.333, Lanzhou, 730050 Gansu China
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
Quantitative Targeted Absolute Proteomics for Better Characterization of an In Vitro Human Blood-Brain Barrier Model Derived from Hematopoietic Stem Cells. Cells 2022; 11:cells11243963. [PMID: 36552728 PMCID: PMC9776576 DOI: 10.3390/cells11243963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
We previously developed an in vitro model of the human blood-brain barrier (BBB) based on the use of endothelial cells derived from CD34+-hematopoietic stem cells and cultured with brain pericytes. The purpose of the present study was to provide information on the protein expression levels of the transporters, receptors, tight junction/adherence junction molecules, and transporter-associated molecules of human brain-like endothelial cells (hBLECs). The absolute protein expression levels were determined by liquid chromatography-mass spectrometry-based quantitative targeted absolute proteomics and compared with those from human brain microvessels (hBMVs). The protein levels of CD144, CD147, MRP4, Annexin A6 and caveolin-1 showed more than 3-fold abundance in hBLECs, those of MCT1, Connexin 43, TfR1, and claudin-5 showed less than 3-fold differences, and the protein levels of other drug efflux transporters and nutrient transporters were less represented in hBLECs than in hBMVs. It is noteworthy that BCRP was more expressed than MDR1 in hBLECs, as this was the case for hBMVs. These results suggest that transports mediated by MCT1, TfR1, and claudin-5-related tight junction function reflect the in vivo BBB situation. The present study provided a better characterization of hBLECs and clarified the equivalence of the transport characteristics between in vitro BBB models and in vivo BBB models using LC-MS/MS-based protein quantification.
Collapse
|
40
|
Naranjo O, Torices S, Clifford PR, Daftari MT, Osborne OM, Fattakhov N, Toborek M. Pericyte infection by HIV-1: a fatal attraction. Retrovirology 2022; 19:27. [PMID: 36476484 PMCID: PMC9730689 DOI: 10.1186/s12977-022-00614-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
While HIV-1 is primarily an infection of CD4 + T cells, there is an emerging interest towards understanding how infection of other cell types can contribute to HIV-associated comorbidities. For HIV-1 to cross from the blood stream into tissues, the virus must come in direct contact with the vascular endothelium, including pericytes that envelope vascular endothelial cells. Pericytes are multifunctional cells that have been recognized for their essential role in angiogenesis, vessel maintenance, and blood flow rate. Most importantly, recent evidence has shown that pericytes can be a target of HIV-1 infection and support an active stage of the viral life cycle, with latency also suggested by in vitro data. Pericyte infection by HIV-1 has been confirmed in the postmortem human brains and in lungs from SIV-infected macaques. Moreover, pericyte dysfunction has been implicated in a variety of pathologies ranging from ischemic stroke to diabetes, which are common comorbidities among people with HIV-1. In this review, we discuss the role of pericytes during HIV-1 infection and their contribution to the progression of HIV-associated comorbidities.
Collapse
Affiliation(s)
- Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Paul R. Clifford
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Manav T. Daftari
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Olivia M. Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Nikolai Fattakhov
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 11336 USA
| |
Collapse
|
41
|
Asah S, Alganem K, McCullumsmith RE, O'Donovan SM. A bioinformatic inquiry of the EAAT2 interactome in postmortem and neuropsychiatric datasets. Schizophr Res 2022; 249:38-46. [PMID: 32197935 PMCID: PMC7494586 DOI: 10.1016/j.schres.2020.03.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Altered expression and localization of the glutamate transporter EAAT2 is found in schizophrenia and other neuropsychiatric (major depression, MDD) and neurological disorders (amyotrophic lateral sclerosis, ALS). However, the EAAT2 interactome, the network of proteins that physically or functionally interact with EAAT2 to support its activity, has yet to be characterized in severe mental illness. We compiled a list of "core" EAAT2 interacting proteins. Using Kaleidoscope, an R-shiny application, we data mined publically available postmortem transcriptome datasets to determine whether components of the EAAT2 interactome are differentially expressed in schizophrenia and, using Reactome, identify which interactome-associated biological pathways are altered. Overall, these "look up" studies highlight region-specific, primarily frontal cortex (dorsolateral prefrontal cortex and anterior cingulate cortex), changes in the EAAT2 interactome and implicate altered metabolism pathways in schizophrenia. Pathway analyses also suggest that perturbation of components of the EAAT2 interactome in animal models of antipsychotic administration impact metabolism. Similar changes in metabolism pathways are seen in ALS, in addition to altered expression of many components of the EAAT2 interactome. However, although EAAT2 expression is altered in a postmortem MDD dataset, few other components of the EAAT2 interactome are changed. Thus, "look up" studies suggest region- and disease-relevant biological pathways related to the EAAT2 interactome that implicate glutamate reuptake perturbations in schizophrenia, while providing a useful tool to exploit "omics" datasets.
Collapse
Affiliation(s)
- Sophie Asah
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Khaled Alganem
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | | | | |
Collapse
|
42
|
Naranjo O, Osborne O, Torices S, Toborek M. In Vivo Targeting of the Neurovascular Unit: Challenges and Advancements. Cell Mol Neurobiol 2022; 42:2131-2146. [PMID: 34086179 PMCID: PMC9056891 DOI: 10.1007/s10571-021-01113-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022]
Abstract
The blood-brain barrier (BBB) is essential for the homeostasis of the central nervous system (CNS). Functions of the BBB are performed by the neurovascular unit (NVU), which consists of endothelial cells, pericytes, astrocytes, microglia, basement membrane, and neurons. NVU cells interact closely and together are responsible for neurovascular coupling, BBB integrity, and transendothelial fluid transport. Studies have shown that NVU dysfunction is implicated in several acute and chronic neurological diseases, including Alzheimer's disease, multiple sclerosis, and stroke. The mechanisms of NVU disruption remain poorly understood, partially due to difficulties in selective targeting of NVU cells. In this review, we discuss the relative merits of available protein markers and drivers of the NVU along with recent advancements that have been made in the field to increase efficiency and specificity of NVU research.
Collapse
Affiliation(s)
- Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Olivia Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland.
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Gautier Bldg., Room 528, 1011 NW 15th Street, Miami, FL, 33136, USA.
| |
Collapse
|
43
|
Kaul D, Schwab SG, Mechawar N, Ooi L, Matosin N. Alterations in Astrocytic Regulation of Excitation and Inhibition by Stress Exposure and in Severe Psychopathology. J Neurosci 2022; 42:6823-6834. [PMID: 38377014 PMCID: PMC9463979 DOI: 10.1523/jneurosci.2410-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022] Open
Abstract
Dysregulation of excitatory and inhibitory signaling is commonly observed in major psychiatric disorders, including schizophrenia, depression, and bipolar disorder, and is often targeted by psychological and pharmacological treatment methods. The balance of excitation and inhibition is highly sensitive to severe psychological stress, one of the strongest risk factors for psychiatric disorders. The role of astrocytes in regulating excitatory and inhibitory signaling is now widely recognized; however, the specific involvement of astrocytes in the context of psychiatric disorders with a history of significant stress exposure remains unclear. In this review, we summarize how astrocytes regulate the balance of excitation and inhibition in the context of stress exposure and severe psychopathology, with a focus on the PFC, a brain area highly implicated in psychopathology. We first focus on preclinical models to demonstrate that the duration of stress (particularly acute vs chronic stress) is key to shaping astrocyte function and downstream behavior. We then provide a hypothesis for how astrocytes are involved in stress-associated cortical signaling imbalance, discuss how this directly contributes to phenotypes of psychopathologies, and provide suggestions for future research. We highlight that astrocytes are a key target to understand and treat the dysregulation of cortical signaling associated with stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Dominic Kaul
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, 2522, Australia
- Molecular Horizons, School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia
| | - Sibylle G Schwab
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, 2522, Australia
- Molecular Horizons, School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia
- School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia
| | - Naguib Mechawar
- Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec H4H 1R3, Canada
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, 2522, Australia
- Molecular Horizons, School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia
| | - Natalie Matosin
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, 2522, Australia
- Molecular Horizons, School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, 2522, Australia
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, Munich, 80804, Germany
| |
Collapse
|
44
|
Fedorenko OY, Paderina DZ, Kornetova EG, Poltavskaya EG, Pozhidaev IV, Goncharova AA, Freidin MB, Bocharova AV, Bokhan NA, Loonen AJM, Ivanova SA. Genes of the Glutamatergic System and Tardive Dyskinesia in Patients with Schizophrenia. Diagnostics (Basel) 2022; 12:diagnostics12071521. [PMID: 35885427 PMCID: PMC9322868 DOI: 10.3390/diagnostics12071521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Tardive dyskinesia (TD) is an extrapyramidal side effect of the long-term use of antipsychotics. In the present study, the role of glutamatergic system genes in the pathogenesis of total TD, as well as two phenotypic forms, orofacial TD and limb-truncal TD, was studied. Methods: A set of 46 SNPs of the glutamatergic system genes (GRIN2A, GRIN2B, GRIK4, GRM3, GRM7, GRM8, SLC1A2, SLC1A3, SLC17A7) was studied in a population of 704 Caucasian patients with schizophrenia. Genotyping was performed using the MassARRAY Analyzer 4 (Agena Bioscience™). Logistic regression analysis was performed to test for the association of TD with the SNPs while adjusting for confounders. Results: No statistically significant associations between the SNPs and TD were found after adjusting for multiple testing. Since three SNPs of the SLC1A2 gene demonstrated nominally significant associations, we carried out a haplotype analysis for these SNPs. This analysis identified a risk haplotype for TD comprising CAT alleles of the SLC1A2 gene SNPs rs1042113, rs10768121, and rs12361171. Nominally significant associations were identified for SLC1A3 rs2229894 and orofacial TD, as well as for GRIN2A rs7192557 and limb-truncal TD. Conclusions: Genes encoding for mGlu3, EAAT2, and EAAT1 may be involved in the development of TD in schizophrenia patients.
Collapse
Affiliation(s)
- Olga Yu. Fedorenko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (O.Y.F.); (D.Z.P.); (E.G.K.); (E.G.P.); (I.V.P.); (A.A.G.); (N.A.B.); (S.A.I.)
| | - Diana Z. Paderina
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (O.Y.F.); (D.Z.P.); (E.G.K.); (E.G.P.); (I.V.P.); (A.A.G.); (N.A.B.); (S.A.I.)
| | - Elena G. Kornetova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (O.Y.F.); (D.Z.P.); (E.G.K.); (E.G.P.); (I.V.P.); (A.A.G.); (N.A.B.); (S.A.I.)
- Department of Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, 634050 Tomsk, Russia
| | - Evgeniya G. Poltavskaya
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (O.Y.F.); (D.Z.P.); (E.G.K.); (E.G.P.); (I.V.P.); (A.A.G.); (N.A.B.); (S.A.I.)
| | - Ivan V. Pozhidaev
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (O.Y.F.); (D.Z.P.); (E.G.K.); (E.G.P.); (I.V.P.); (A.A.G.); (N.A.B.); (S.A.I.)
| | - Anastasiia A. Goncharova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (O.Y.F.); (D.Z.P.); (E.G.K.); (E.G.P.); (I.V.P.); (A.A.G.); (N.A.B.); (S.A.I.)
| | - Maxim B. Freidin
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (M.B.F.); (A.V.B.)
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Anna V. Bocharova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (M.B.F.); (A.V.B.)
| | - Nikolay A. Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (O.Y.F.); (D.Z.P.); (E.G.K.); (E.G.P.); (I.V.P.); (A.A.G.); (N.A.B.); (S.A.I.)
- Department of Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, 634050 Tomsk, Russia
| | - Anton J. M. Loonen
- Unit of PharmacoTherapy, Epidemiology & Economics, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands
- Correspondence:
| | - Svetlana A. Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia; (O.Y.F.); (D.Z.P.); (E.G.K.); (E.G.P.); (I.V.P.); (A.A.G.); (N.A.B.); (S.A.I.)
- Department of Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, 634050 Tomsk, Russia
| |
Collapse
|
45
|
Sijben HJ, Dall’ Acqua L, Liu R, Jarret A, Christodoulaki E, Onstein S, Wolf G, Verburgt SJ, Le Dévédec SE, Wiedmer T, Superti-Furga G, IJzerman AP, Heitman LH. Impedance-Based Phenotypic Readout of Transporter Function: A Case for Glutamate Transporters. Front Pharmacol 2022; 13:872335. [PMID: 35677430 PMCID: PMC9169222 DOI: 10.3389/fphar.2022.872335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/29/2022] [Indexed: 11/18/2022] Open
Abstract
Excitatory amino acid transporters (EAAT/SLC1) mediate Na+-dependent uptake of extracellular glutamate and are potential drug targets for neurological disorders. Conventional methods to assess glutamate transport in vitro are based on radiolabels, fluorescent dyes or electrophysiology, which potentially compromise the cell’s physiology and are generally less suited for primary drug screens. Here, we describe a novel label-free method to assess human EAAT function in living cells, i.e., without the use of chemical modifications to the substrate or cellular environment. In adherent HEK293 cells overexpressing EAAT1, stimulation with glutamate or aspartate induced cell spreading, which was detected in real-time using an impedance-based biosensor. This change in cell morphology was prevented in the presence of the Na+/K+-ATPase inhibitor ouabain and EAAT inhibitors, which suggests the substrate-induced response was ion-dependent and transporter-specific. A mechanistic explanation for the phenotypic response was substantiated by actin cytoskeleton remodeling and changes in the intracellular levels of the osmolyte taurine, which suggests that the response involves cell swelling. In addition, substrate-induced cellular responses were observed for cells expressing other EAAT subtypes, as well as in a breast cancer cell line (MDA-MB-468) with endogenous EAAT1 expression. These findings allowed the development of a label-free high-throughput screening assay, which could be beneficial in early drug discovery for EAATs and holds potential for the study of other transport proteins that modulate cell shape.
Collapse
Affiliation(s)
- Hubert J. Sijben
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Laura Dall’ Acqua
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Rongfang Liu
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Abigail Jarret
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Eirini Christodoulaki
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Svenja Onstein
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Gernot Wolf
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Simone J. Verburgt
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Sylvia E. Le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Adriaan P. IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Laura H. Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
- Oncode Institute, Leiden, Netherlands
- *Correspondence: Laura H. Heitman,
| |
Collapse
|
46
|
Host cell proteins modulated upon Toxoplasma infection identified using proteomic approaches: a molecular rationale. Parasitol Res 2022; 121:1853-1865. [PMID: 35552534 DOI: 10.1007/s00436-022-07541-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
Toxoplasma gondii is a pathogenic protozoan parasite belonging to the apicomplexan phylum that infects the nucleated cells of warm-blooded hosts leading to an infectious disease known as toxoplasmosis. Apicomplexan parasites such as T. gondii can display different mechanisms to control or manipulate host cells signaling at different levels altering the host subcellular genome and proteome. Indeed, Toxoplasma is able to modulate host cell responses (especially immune responses) during infection to its advantage through both structural and functional changes in the proteome of different infected cells. Consequently, parasites can transform the invaded cells into a suitable environment for its own replication and the induction of infection. Proteomics as an applicable tool can identify such critical proteins involved in pathogen (Toxoplasma)-host cell interactions and consequently clarify the cellular mechanisms that facilitate the entry of pathogens into host cells, and their replication and transmission, as well as the central mechanisms of host defense against pathogens. Accordingly, the current paper reviews several proteins (identified using proteomic approaches) differentially expressed in the proteome of Toxoplasma-infected host cells (macrophages and human foreskin fibroblasts) and tissues (brain and liver) and highlights their plausible functions in the cellular biology of the infected cells. The identification of such modulated proteins and their related cell impact (cell responses/signaling) can provide further information regarding parasite pathogenesis and biology that might lead to a better understanding of therapeutic strategies and novel drug targets.
Collapse
|
47
|
Beeraka NM, Vikram PRH, Greeshma MV, Uthaiah CA, Huria T, Liu J, Kumar P, Nikolenko VN, Bulygin KV, Sinelnikov MY, Sukocheva O, Fan R. Recent Investigations on Neurotransmitters' Role in Acute White Matter Injury of Perinatal Glia and Pharmacotherapies-Glia Dynamics in Stem Cell Therapy. Mol Neurobiol 2022; 59:2009-2026. [PMID: 35041139 DOI: 10.1007/s12035-021-02700-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023]
Abstract
Periventricular leukomalacia (PVL) and cerebral palsy are two neurological disease conditions developed from the premyelinated white matter ischemic injury (WMI). The significant pathophysiology of these diseases is accompanied by the cognitive deficits due to the loss of function of glial cells and axons. White matter makes up 50% of the brain volume consisting of myelinated and non-myelinated axons, glia, blood vessels, optic nerves, and corpus callosum. Studies over the years have delineated the susceptibility of white matter towards ischemic injury especially during pregnancy (prenatal, perinatal) or immediately after child birth (postnatal). Impairment in membrane depolarization of neurons and glial cells by ischemia-invoked excitotoxicity is mediated through the overactivation of NMDA receptors or non-NMDA receptors by excessive glutamate influx, calcium, or ROS overload and has been some of the well-studied molecular mechanisms conducive to the injury of white matter. Expression of glutamate receptors (GluR) and transporters (GLT1, EACC1, and GST) has significant influence in glial and axonal-mediated injury of premyelinated white matter during PVL and cerebral palsy. Predominantly, the central premyelinated axons express extensive levels of functional NMDA GluR receptors to confer ischemic injury to premyelinated white matter which in turn invoke defects in neural plasticity. Several underlying molecular mechanisms are yet to be unraveled to delineate the complete pathophysiology of these prenatal neurological diseases for developing the novel therapeutic modalities to mitigate pathophysiology and premature mortality of newborn babies. In this review, we have substantially discussed the above multiple pathophysiological aspects of white matter injury along with glial dynamics, and the pharmacotherapies including recent insights into the application of MSCs as therapeutic modality in treating white matter injury.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - P R Hemanth Vikram
- Department of Pharmaceutical Chemistry, JSS Pharmacy College, Mysuru, Karnataka, India
| | - M V Greeshma
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
| | - Chinnappa A Uthaiah
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
| | - Tahani Huria
- Faculty of Medicine, Benghazi University, Benghazi, Libya
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, LE1 7RH, UK
| | - Junqi Liu
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China
| | - Pramod Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER-Guwahati), SilaKatamur (Halugurisuk), Changsari, Kamrup, 781101, Assam, India
| | - Vladimir N Nikolenko
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
- Department of Normal and Topographic Anatomy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill V Bulygin
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Mikhail Y Sinelnikov
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
- Research Institute of Human Morphology, 3 Tsyurupy Street, Moscow, 117418, Russian Federation
| | - Olga Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Ruitai Fan
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China.
| |
Collapse
|
48
|
Nicholson S, Baccarelli A, Prada D. Role of brain extracellular vesicles in air pollution-related cognitive impairment and neurodegeneration. ENVIRONMENTAL RESEARCH 2022; 204:112316. [PMID: 34728237 PMCID: PMC8671239 DOI: 10.1016/j.envres.2021.112316] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 09/15/2021] [Accepted: 10/28/2021] [Indexed: 05/07/2023]
Abstract
A relationship between environmental exposure to air pollution and cognitive impairment and neurological disorders has been described. Previous literature has focused on the direct effects of the air pollution components on neuronal and glial cells, as well as on involvement of oxidative stress and neuroinflammation on microglia and astrocyte reactivity. However, other mechanisms involved in the air pollution effects on central nervous system (CNS) toxicity can be playing critical roles. Increasingly, extracellular vesicle's (EVs) mediated intercellular communication is being recognized as impacting the development of cognitive impairment and neurological disorders like Alzheimer's disease and others. Here we describe the available evidence about toxic air pollutants and its components on brain, an involvement of brain cells specific and EVs types (based in the origin or in the size of EVs) in the initiation, exacerbation, and propagation of the neurotoxic effects (inflammation, neurodegeneration, and accumulation of neurotoxic proteins) induced by air pollution in the CNS. Additionally, we discuss the identification and isolation of neural-derived EVs from human plasma, the most common markers for neural-derived EVs, and their potential for use as diagnostic or therapeutic molecules for air pollution-related cognitive impairment and neurodegeneration.
Collapse
Affiliation(s)
- Stacia Nicholson
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA
| | - Diddier Prada
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA; Instituto Nacional de Cancerología, Mexico City, 14080, Mexico.
| |
Collapse
|
49
|
Rimmele TS, Li S, Andersen JV, Westi EW, Rotenberg A, Wang J, Aldana BI, Selkoe DJ, Aoki CJ, Dulla CG, Rosenberg PA. Neuronal Loss of the Glutamate Transporter GLT-1 Promotes Excitotoxic Injury in the Hippocampus. Front Cell Neurosci 2022; 15:788262. [PMID: 35035352 PMCID: PMC8752461 DOI: 10.3389/fncel.2021.788262] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022] Open
Abstract
GLT-1, the major glutamate transporter in the mammalian central nervous system, is expressed in presynaptic terminals that use glutamate as a neurotransmitter, in addition to astrocytes. It is widely assumed that glutamate homeostasis is regulated primarily by glutamate transporters expressed in astrocytes, leaving the function of GLT-1 in neurons relatively unexplored. We generated conditional GLT-1 knockout (KO) mouse lines to understand the cell-specific functions of GLT-1. We found that stimulus-evoked field extracellular postsynaptic potentials (fEPSPs) recorded in the CA1 region of the hippocampus were normal in the astrocytic GLT-1 KO but were reduced and often absent in the neuronal GLT-1 KO at 40 weeks. The failure of fEPSP generation in the neuronal GLT-1 KO was also observed in slices from 20 weeks old mice but not consistently from 10 weeks old mice. Using an extracellular FRET-based glutamate sensor, we found no difference in stimulus-evoked glutamate accumulation in the neuronal GLT-1 KO, suggesting a postsynaptic cause of the transmission failure. We hypothesized that excitotoxicity underlies the failure of functional recovery of slices from the neuronal GLT-1 KO. Consistent with this hypothesis, the non-competitive NMDA receptor antagonist MK801, when present in the ACSF during the recovery period following cutting of slices, promoted full restoration of fEPSP generation. The inclusion of an enzymatic glutamate scavenging system in the ACSF conferred partial protection. Excitotoxicity might be due to excess release or accumulation of excitatory amino acids, or to metabolic perturbation resulting in increased vulnerability to NMDA receptor activation. Previous studies have demonstrated a defect in the utilization of glutamate by synaptic mitochondria and aspartate production in the synGLT-1 KO in vivo, and we found evidence for similar metabolic perturbations in the slice preparation. In addition, mitochondrial cristae density was higher in synaptic mitochondria in the CA1 region in 20–25 weeks old synGLT-1 KO mice in the CA1 region, suggesting compensation for loss of axon terminal GLT-1 by increased mitochondrial efficiency. These data suggest that GLT-1 expressed in presynaptic terminals serves an important role in the regulation of vulnerability to excitotoxicity, and this regulation may be related to the metabolic role of GLT-1 expressed in glutamatergic axon terminals.
Collapse
Affiliation(s)
- Theresa S Rimmele
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jens Velde Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Emil W Westi
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Rotenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| | - Jianlin Wang
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Blanca Irene Aldana
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Chiye J Aoki
- Center for Neural Science, New York University, NY, United States.,Neuroscience Institute NYU Langone Medical Center, NY, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Paul Allen Rosenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
50
|
Nguyen YTK, Ha HTT, Nguyen TH, Nguyen LN. The role of SLC transporters for brain health and disease. Cell Mol Life Sci 2021; 79:20. [PMID: 34971415 PMCID: PMC11071821 DOI: 10.1007/s00018-021-04074-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/05/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022]
Abstract
The brain exchanges nutrients and small molecules with blood via the blood-brain barrier (BBB). Approximately 20% energy intake for the body is consumed by the brain. Glucose is known for its critical roles for energy production and provides substrates for biogenesis in neurons. The brain takes up glucose via glucose transporters GLUT1 and 3, which are expressed in several neural cell types. The brain is also equipped with various transport systems for acquiring amino acids, lactate, ketone bodies, lipids, and cofactors for neuronal functions. Unraveling the mechanisms by which the brain takes up and metabolizes these nutrients will be key in understanding the nutritional requirements in the brain. This could also offer opportunities for therapeutic interventions in several neurological disorders. For instance, emerging evidence suggests a critical role of lactate as an alternative energy source for neurons. Neuronal cells express monocarboxylic transporters to acquire lactate. As such, treatment of GLUT1-deficient patients with ketogenic diets to provide the brain with alternative sources of energy has been shown to improve the health of the patients. Many transporters are present in the brain, but only a small number has been characterized. In this review, we will discuss about the roles of solute carrier (SLC) transporters at the blood brain barrier (BBB) and neural cells, in transport of nutrients and metabolites in the brain.
Collapse
Affiliation(s)
- Yen T K Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Hoa T T Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Tra H Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
- SLING/Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- Immunology Translational and Cardiovascular Disease Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.
| |
Collapse
|