1
|
Liu XH, Zhang LY, Liu XY, Zhang JG, Hu YY, Zhao CG, Xian XH, Li WB, Zhang M. Transformation of A1/A2 Astrocytes Participates in Brain Ischemic Tolerance Induced by Cerebral Ischemic Preconditioning via Inhibiting NDRG2. Neurochem Res 2024; 49:1665-1676. [PMID: 38411782 DOI: 10.1007/s11064-024-04134-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Cerebral ischemic preconditioning (CIP) has been shown to improve brain ischemic tolerance against subsequent lethal ischemia. Reactive astrocytes play important roles in cerebral ischemia-reperfusion. Recent studies have shown that reactive astrocytes can be polarized into neurotoxic A1 phenotype (C3d) and neuroprotective A2 phenotype (S100A10). However, their role in CIP remains unclear. Here, we focused on the role of N-myc downstream-regulated gene 2 (NDRG2) in regulating the transformation of A1/A2 astrocytes and promoting to brain ischemic tolerance induced by CIP. A Sprague Dawley rat model of middle cerebral artery occlusion/reperfusion (MCAO/R) was used. Rats were divided into the following six groups: (1) sham group; (2) CIP group: left middle cerebral artery was blocked for 10 min; (3) MCAO/R group: left middle cerebral artery was blocked for 90 min; (4) CIP + MCAO/R group: CIP was performed 72 h before MCAO/R; (5) AAV-NDRG2 + CIP + MCAO/R group: adeno-associated virus (AAV) carrying NDRG2 was administered 14 days before CIP + MCAO/R; (6) AAV-Ctrl + CIP + MCAO/R group: empty control group. The rats were subjected to neurological evaluation 24 h after the above treatments, and then were sacrificed for 2, 3, 5-triphenyltetraolium chloride staining, thionin staining, immunofluorescence and western blot analysis. In CIP + MCAO/R group, the neurological deficit scores decreased, infarct volume reduced, and neuronal density increased compared with MCAO/R group. Notably, CIP significantly increased S100A10 expression and the number of S100A10+/GFAP+ cells, and also increased NDRG2 expression. MCAO/R significantly decreased S100A10 expression and the number of S100A10+/GFAP+ cells yet increased C3d expression and the number of C3d+/GFAP+ cells and NDRG2 expression, and these trends were reversed by CIP + MCAO/R. Furthermore, over-expression of NDRG2 before CIP + MCAO/R, the C3d expression and the number of C3d+/GFAP+ cells increased, while S100A10 expression and the number of S100A10+/GFAP+ cells decreased. Meanwhile, over-expression of NDRG2 blocked the CIP-induced brain ischemic tolerance. Taken together, these results suggest that CIP exerts neuroprotective effects against ischemic injury by suppressing A1 astrocyte polarization and promoting A2 astrocyte polarization via inhibiting NDRG2 expression.
Collapse
Affiliation(s)
- Xiao-Hua Liu
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Department of Physiology, Shijiazhuang Medical College, Shijiazhuang, 050000, People's Republic of China
| | - Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China
| | - Xi-Yun Liu
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China.
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China
| | - Chen-Guang Zhao
- Department of foreign language, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050017, People's Republic of China.
| |
Collapse
|
2
|
Liu XY, Zhang LY, Wang XY, Li SC, Hu YY, Zhang JG, Xian XH, Li WB, Zhang M. STAT4-Mediated Klotho Up-Regulation Contributes to the Brain Ischemic Tolerance by Cerebral Ischemic Preconditioning via Inhibiting Neuronal Pyroptosis. Mol Neurobiol 2024; 61:2336-2356. [PMID: 37875707 DOI: 10.1007/s12035-023-03703-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023]
Abstract
Our previous study has proved that the Klotho up-regulation participated in cerebral ischemic preconditioning (CIP)-induced brain ischemic tolerance. However, the exact neuroprotective mechanism of Klotho in CIP remains unclear. We explored the hypothesis that STAT4-mediated Klotho up-regulation contributes to the CIP-induced brain ischemic tolerance via inhibiting neuronal pyroptosis. Firstly, the expressions of pyroptosis-associated proteins (i.e., NLRP3, GSDMD, pro-caspase-1, and cleaved caspase-1) in hippocampal CA1 region were determined during the process of brain ischemic tolerance. We found the expression of pyroptosis-associated proteins was significantly up-regulated in the ischemic insult (II) group, and showed no significant changes in the CIP group. The expression level of each pyroptosis-associated proteins was lower in the CIP + II group than that in the II group. Inhibition of Klotho expression increased the expression of pyroptosis-associated proteins in the CIP + II group and blocked the CIP-induced brain ischemic tolerance. Injection of Klotho protein decreased the expression of pyroptosis-associated proteins in the II group, and protected neurons from ischemic injury. Secondly, the transcription factor STAT4 of Klotho was identified by bioinformatic analysis. Double luciferase reporter gene assay and chromatin immunoprecipitation assay showed STAT4 can bind to the site between nt - 881 and - 868 on the Klotho promoter region and positively regulates Klotho expression. Moreover, we found CIP significantly enhanced the expression of STAT4. Knockdown STAT4 suppressed Klotho up-regulation after CIP and blocked the CIP-induced brain ischemic tolerance. Collectively, it can be concluded that STAT4-mediated the up-regulation of Klotho contributed to the brain ischemic tolerance induced by CIP via inhibiting pyroptosis.
Collapse
Affiliation(s)
- Xi-Yun Liu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
| | - Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, People's Republic of China
| | - Xiao-Yu Wang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
| | - Shi-Chao Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei, 050017, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, People's Republic of China.
| |
Collapse
|
3
|
Končeková J, Kotorová K, Gottlieb M, Bona M, Bonová P. Changes in excitatory amino acid transporters in response to remote ischaemic preconditioning and glutamate excitotoxicity. Neurochem Int 2024; 173:105658. [PMID: 38135159 DOI: 10.1016/j.neuint.2023.105658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/22/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
The successful implementation of remote ischaemic conditioning as a clinical neuroprotective strategy requires a thorough understanding of its basic principles, which can be modified for each patient. The mechanisms of glutamate homeostasis appear to be a key component. In the current study, we focused on the brain-to-blood glutamate shift mediated by glutamate transporters (excitatory amino acid transports [EAATs]) and the effect of remote ischaemic preconditioning (RIPC) as a mediator of ischaemic tolerance. We used model mimicking ischaemia-mediated excitotoxicity (intracerebroventricular administration of glutamate) to avoid the indirect effect of ischaemia-triggered mechanisms. We found quantitative changes in EAAT2 and EAAT3 and altered membrane trafficking of EAAT1 on the cells of the choroid plexus. These changes could underlie the beneficial effects of ischaemic tolerance. There was reduced oxidative stress and increased glutathione level after RIPC treatment. Moreover, we determined the stimulus-specific response on EAATs. While glutamate overdose stimulated EAAT2 and EAAT3 overexpression, RIPC induced membrane trafficking of EAAT1 and EAAT2 rather than a change in their expression. Taken together, mechanisms related to glutamate homeostasis, especially EAAT-mediated transport, represents a powerful tool of ischaemic tolerance and allow a certain amount of flexibility based on the stimulus used.
Collapse
Affiliation(s)
- Jana Končeková
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, Soltesovej 4-6, Košice, 040 01, Slovak Republic
| | - Klaudia Kotorová
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, Soltesovej 4-6, Košice, 040 01, Slovak Republic
| | - Miroslav Gottlieb
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, Soltesovej 4-6, Košice, 040 01, Slovak Republic
| | - Martin Bona
- Department of Medical Physiology, Faculty of Medicine, University of Pavol Jozef Safarik, Košice, 040 01, Slovak Republic
| | - Petra Bonová
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, Soltesovej 4-6, Košice, 040 01, Slovak Republic.
| |
Collapse
|
4
|
Huang S, Ren C, Luo Y, Ding Y, Ji X, Li S. New insights into the roles of oligodendrocytes regulation in ischemic stroke recovery. Neurobiol Dis 2023:106200. [PMID: 37321419 DOI: 10.1016/j.nbd.2023.106200] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/20/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023] Open
Abstract
Oligodendrocytes (OLs), the myelin-forming cells of the central nervous system, are integral to axonal integrity and function. Hypoxia-ischemia episodes can cause severe damage to these vulnerable cells through excitotoxicity, oxidative stress, inflammation, and mitochondrial dysfunction, leading to axonal dystrophy, neuronal dysfunction, and neurological impairments. OLs damage can result in demyelination and myelination disorders, severely impacting axonal function, structure, metabolism, and survival. Adult-onset stroke, periventricular leukomalacia, and post-stroke cognitive impairment primarily target OLs, making them a critical therapeutic target. Therapeutic strategies targeting OLs, myelin, and their receptors should be given more emphasis to attenuate ischemia injury and establish functional recovery after stroke. This review summarizes recent advances on the function of OLs in ischemic injury, as well as the present and emerging principles that serve as the foundation for protective strategies against OL deaths.
Collapse
Affiliation(s)
- Shuangfeng Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China; Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yumin Luo
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China; Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Sijie Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China; Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Beresewicz-Haller M. Hippocampal region-specific endogenous neuroprotection as an approach in the search for new neuroprotective strategies in ischemic stroke. Fiction or fact? Neurochem Int 2023; 162:105455. [PMID: 36410452 DOI: 10.1016/j.neuint.2022.105455] [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: 06/30/2022] [Revised: 11/03/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Ischemic stroke is the leading cause of death and long-term disability worldwide, and, while considerable progress has been made in understanding its pathophysiology, the lack of effective treatments remains a major concern. In that context, receiving more and more consideration as a promising therapeutic method is the activation of natural adaptive mechanisms (endogenous neuroprotection) - an approach that seeks to enhance and/or stimulate the endogenous processes of plasticity and protection of the neuronal system that trigger the brain's intrinsic capacity for self-defence. Ischemic preconditioning is a classic example of endogenous neuroprotection, being the process by which one or more brief, non-damaging episodes of ischemia-reperfusion (I/R) induce tissue resistance to subsequent prolonged, damaging ischemia. Another less-known example is resistance to an I/R episode mounted by the hippocampal region consisting of CA2, CA3, CA4 and the dentate gyrus (here abbreviated to CA2-4, DG). This can be contrasted with the ischemia-vulnerable CA1 region. There is not yet a good understanding of these different sensitivities of the hippocampal regions, and hence of the endogenous neuroprotection characteristic of CA2-4, DG. However, this region is widely reported to have properties distinct from CA1, and capable of generating resistance to an I/R episode. These include activation of neurotrophic and neuroprotective factors, greater activation of anti-excitotoxic and anti-oxidant mechanisms, increased plasticity potential, a greater energy reserve and improved mitochondrial function. This review seeks to summarize properties of CA2-4, DG in the context of endogenous neuroprotection, and then to assess the potential utility of these properties to therapeutic approaches. In so doing, it appears to represent the first such addressing of the issue of ischemia resistance attributable to CA2-4, DG.
Collapse
|
6
|
Koizumi S, Hirayama Y. Ischemic Tolerance Induced by Glial Cells. Neurochem Res 2022; 47:2522-2528. [PMID: 35920970 PMCID: PMC9463280 DOI: 10.1007/s11064-022-03704-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/09/2022] [Accepted: 07/16/2022] [Indexed: 11/05/2022]
Abstract
Ischemic tolerance is a phenomenon in which resistance to subsequent invasive ischemia is acquired by a preceding noninvasive ischemic application, and is observed in many organs, including the brain, the organ most vulnerable to ischemic insult. To date, much research has been conducted on cerebral ischemic tolerance as a cell-autonomous action of neurons. In this article, we review the essential roles of microglia and astrocytes in the acquisition of ischemic tolerance through neuron-non-autonomous mechanisms, where the two types of glial cells function in a concerted manner to induce ischemic tolerance.
Collapse
Affiliation(s)
- Schuichi Koizumi
- Department of Neuropharmacology, Yamanashi, Japan. .,Yamanashi GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 409-3898, Yamanashi, Japan.
| | - Yuri Hirayama
- Department of Neuropharmacology, Yamanashi, Japan.,Department of Pharmacology, Graduate School of Medicine, Chiba University, 260-8670, Chiba, Japan
| |
Collapse
|
7
|
Li S, Zhang L, Lin J, Su A, Liu X, Zhang J, Xian X, Hu Y, Li W, Sun S, Zhang M. LncRNA BIRF Promotes Brain Ischemic Tolerance Induced By Cerebral Ischemic Preconditioning Through Upregulating GLT-1 via Sponging miR-330-5p. Mol Neurobiol 2022; 59:3996-4014. [PMID: 35451738 PMCID: PMC9167204 DOI: 10.1007/s12035-022-02841-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/04/2022] [Indexed: 10/24/2022]
Abstract
Long noncoding RNAs (lncRNAs) play an important regulatory role in various diseases. However, the role of lncRNAs in brain ischemic tolerance (BIT) induced by cerebral ischemic preconditioning (CIPC) is still unknown. The lncRNA profile of rat cortical astrocytes pretreated with ischemic preconditioning was analyzed by high-throughput sequencing. The results of Cell-Counting Kit-8 (CCK-8) assay showed that a novel lncRNA, NONRATT009133.2, which we referred to as brain ischemia-related factor (BIRF), was highly correlated with BIT. Through bioinformatics analysis, we predicted that BIRF, miR-330-5p, and GLT-1 (also named Slc1a2) might constitute a ceRNA regulatory network in the induction of BIT. We found that BIRF was upregulated by CIPC, which promoted GLT-1 expression and BIT induction. BIRF could directly bind to miR-330-5p. Furthermore, miR-330-5p directly targeted GLT-1, and miR-330-5p inhibited both GLT-1 expression and BIT induction in vitro and in vivo. Moreover, BIRF acts as a molecular sponge to competitively bind to miR-330-5p with GLT-1 mRNA, while the miR-330-5p inhibitor reversed all the effects of BIRF siRNA on GLT-1 expression and neuronal vitality. Taken together, our results demonstrate the important roles of the BIRF/miR-330-5p/GLT-1 axis in the induction of BIT by CIPC. BIRF may be a potentially effective therapeutic strategy against stroke injury.
Collapse
Affiliation(s)
- Shichao Li
- Key Laboratory of Critical Disease Mechanism and intervention of Hebei Province, Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Lingyan Zhang
- Key Laboratory of Critical Disease Mechanism and intervention of Hebei Province, Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Jiajie Lin
- Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Department of Biochemistry and Molecular Biology, Cardiovascular Medical Science Center, Shijiazhuang, China
| | - Achou Su
- Key Laboratory of Critical Disease Mechanism and intervention of Hebei Province, Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Xiyun Liu
- Key Laboratory of Critical Disease Mechanism and intervention of Hebei Province, Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Jingge Zhang
- Key Laboratory of Critical Disease Mechanism and intervention of Hebei Province, Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Xiaohui Xian
- Key Laboratory of Critical Disease Mechanism and intervention of Hebei Province, Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Yuyan Hu
- Key Laboratory of Critical Disease Mechanism and intervention of Hebei Province, Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Wenbin Li
- Key Laboratory of Critical Disease Mechanism and intervention of Hebei Province, Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Shaoguang Sun
- Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Department of Biochemistry and Molecular Biology, Cardiovascular Medical Science Center, Shijiazhuang, China.
| | - Min Zhang
- Key Laboratory of Critical Disease Mechanism and intervention of Hebei Province, Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
8
|
Effects of Ozone on Hippocampus BDNF and Fos Expressions in Rats with Chronic Compression of Dorsal Root Ganglia. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5572915. [PMID: 34869766 PMCID: PMC8642004 DOI: 10.1155/2021/5572915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 10/26/2021] [Indexed: 11/30/2022]
Abstract
The effects of ozone on hippocampal expression levels of brain-derived neurotrophic factor (BDNF) and c-fos protein (Fos) were evaluated in rats with chronic compression of dorsal root ganglia (CCD). Forty-eight adult female Sprague-Dawley rats were randomly divided into the following 4 groups (n = 12): sham operation (sham group), CCD group, CCD with 20 μg/ml of ozone (CCD + AO3 group), and CCD with 40 μg/ml of ozone (CCD + BO3 group). Except the sham group, unilateral L5 dorsal root ganglion (DRG) compression was performed on all other groups. On days 1, 2, and 4 after the operation, the CCD + AO3 and CCD + BO3 groups were injected with 100 μl of ozone with concentrations of 20 and 40 μg/ml, respectively. Thermal withdrawal latencies (TWLs) and mechanical withdrawal thresholds (MWTs) were measured at various time points before and after the operation. BDNF and Fos expressions were examined in the extracted hippocampi using immunohistochemistry. The TWLs and MWTs of CCD model rats that received ozone were lower with decreased BDNF and increased Fos expression levels, on day 21 after the operation, compared to those of the sham group (P < 0.05). The TWLs and MWTs of the CCD + AO3 and CCD + BO3 groups were higher with increased BDNF and decreased Fos expression levels, on day 21 after the operation, compared to those of the CCD group (P < 0.05). The TWLs were longer and the MWTs were higher in the CCD + BO3 group at each time point with increased BDNF and decreased Fos expression levels, on day 21 after the operation, compared to those of the CCD + AO3 group (P < 0.05). Our results revealed that ozone can relieve the neuropathic pain caused by the pathological neuralgia resulting from DRG compression in rats. The mechanism of action for ozone is likely associated with changes in BDNF and Fos expression levels in the hippocampus.
Collapse
|
9
|
Sulbactam improves binding property and uptake capacity of glutamate transporter-1 and decreases glutamate concentration in the CA1 region of hippocampus of global brain ischemic rats. Amino Acids 2021; 53:1649-1661. [PMID: 34716803 DOI: 10.1007/s00726-021-03088-3] [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: 08/03/2021] [Accepted: 10/07/2021] [Indexed: 10/19/2022]
Abstract
Glutamate transporter-1 (GLT-1) removes most glutamate in the synaptic cleft. Sulbactam confers neuronal protection against ischemic insults in the hippocampal CA1 region accompanied by the upregulation of GLT-1 expression in rats. The present study further investigates the effect of sulbactam on the binding property and uptake capacity of GLT-1 for glutamate, and the change in extracellular glutamate concentration in the hippocampal CA1 region of rats with global brain ischemia. The binding property and uptake capacity of GLT-1 were measured using a radioligand binding and uptake assay, respectively, with L-3H-glutamate. The extracellular glutamate concentration was detected using microdialysis and high-performance liquid chromatography-mass spectrometry. Neuropathological evaluation was performed based on thionin staining. It was shown that sulbactam pre-treatment changed GLT-1 binding property, including increased Bmax and decreased Kd values, increased GLT-1 uptake capacity for glutamate, and inhibited the elevation of extracellular glutamate concentration in rats with global cerebral ischemia. These effects of sulbactam were accompanied by its neuronal protection on the hippocampal CA1 neurons against delayed neuronal death resulted from ischemic insult. Furthermore, administration of GLT-1 antisense oligodeoxynucleotides, which inhibited the expression of GLT-1, blocked the aforementioned sulbactam-related effects, which suggested that GLT-1 upregulation mediated the above effect although other mechanisms independent of the upregulation of GLT-1 expression could not be excluded. It could be concluded that sulbactam improves the binding property and uptake capacity of GLT-1 for glutamate and then reduces the glutamate concentration and excitotoxicity during global cerebral ischemia, which contributes to the neuroprotection of sulbactam against brain ischemia.
Collapse
|
10
|
Ma D, An Q, Zhang Z, Bian Q, Li Y, Li Y, Zhang S. Head Mild Hypothermia Exerts a Neuroprotective Role in Ischemia–Reperfusion Injury by Maintaining Glial Glutamate Transporter 1. Ther Hypothermia Temp Manag 2021; 11:155-163. [PMID: 32985953 DOI: 10.1089/ther.2020.0012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Dongyang Ma
- Department of Anesthesiology, Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Qi An
- Department of Anesthesiology, Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Zhiqiang Zhang
- Department of Anesthesiology, Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Qinghu Bian
- Department of Anesthesiology, Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yanan Li
- Department of Anesthesiology, Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yanli Li
- Department of Anesthesiology, Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Shan Zhang
- Department of Anesthesiology, Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| |
Collapse
|
11
|
Sun YW, Zhang LY, Gong SJ, Hu YY, Zhang JG, Xian XH, Li WB, Zhang M. The p38 MAPK/NF-κB pathway mediates GLT-1 up-regulation during cerebral ischemic preconditioning-induced brain ischemic tolerance in rats. Brain Res Bull 2021; 175:224-233. [PMID: 34343641 DOI: 10.1016/j.brainresbull.2021.07.029] [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: 06/02/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
Our previous finding suggests that p38 MAPK contributes to the GLT-1 upregulation during induction of brain ischemic tolerance by cerebral ischemic preconditioning (CIP), however, the underlying mechanism is still unclear. Here, we investigated the molecular mechanisms underlying the CIP-induced GLT-1 upregulation by using Western blotting, Co-immunoprecipitation (Co-IP), electrophoretic mobility shift assay (EMSA) and thionin staining in rat hippocampus CA1 subset. We found that application of BAY11-7082 (an inhibitor of NF-κB), or dihydrokainate (an inhibitor of GLT-1), or SB203580 (an inhibitor of p38 MAPK) could attenuate the CIP-induced neuronal protection in hippocampus CA1 region of rats. Moreover, CIP caused rapid activation of NF-κB, as evidenced by nuclear translocation of NF-κB p50 protein, which led to active p50/p65 dimer formation and increased DNA binding activity. GLT-1 was also increased after CIP. Pretreatment with BAY11-7082 blocked the CIP-induced GLT-1 upregulation. The above results suggest that NF-κB participates in GLT-1 up-regulation during the induction of brain ischemic tolerance by CIP. We also found that pretreatment with SB203580 caused significant reduction of NF-κB p50 protein in nucleus, NF-κB p50/p65 dimer nuclear translocation and DNA binding activity of NF-κB. Together, we conclude that p38 MAPK/NF-κB pathway participates in the mediation of GLT-1 up-regulation during the induction of brain ischemic tolerance induced by CIP.
Collapse
Affiliation(s)
- Ya-Wei Sun
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Xing Tai People's Hospital, 16 Hong Xing Road, Xing Tai, 054001, People's Republic of China
| | - Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Shu-Juan Gong
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China.
| |
Collapse
|
12
|
Fan S, Li L, Xian X, Liu L, Gao J, Li W. Ceftriaxone regulates glutamate production and vesicular assembly in presynaptic terminals through GLT-1 in APP/PS1 mice. Neurobiol Learn Mem 2021; 183:107480. [PMID: 34153453 DOI: 10.1016/j.nlm.2021.107480] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 05/13/2021] [Accepted: 06/16/2021] [Indexed: 10/21/2022]
Abstract
Perturbations in the glutamate-glutamine cycle and glutamate release from presynaptic terminals have been involved in the development of cognitive deficits in Alzheimer's disease (AD) patients and mouse models. Glutamate transporter-1 (GLT-1) removes glutamate from the synaptic cleft and transports it into astrocytes, where it is used as substrate for the glutamate-glutamine cycle. Ceftriaxone has been reported to improve cognitive deficits in AD mice by increasing GLT-1 expression, glutamate transformation to glutamine, and glutamine efflux from astrocytes. However, the impact of ceftriaxone on glutamine metabolism in neurons is unknown. The present study aimed to investigate whether ceftriaxone regulated the production and vesicular assembly of glutamate in the presynaptic terminals of neurons and to determine GLT-1 involvement in this process. We used the amyloid precursor protein (APP)/presenilin-1 (PS1) AD mouse model and GLT-1 knockdown APP/PS1 (GLT-1+/-/APP/PS1) mice. The expression levels of sodium-coupled neutral amino-acid transporter 1 (SNAT1) and vesicular glutamate transporters 1 and 2 (VGLUT1/2) were analyzed by immunofluorescence and immunohistochemistry staining as well as by Western blotting. Glutaminase activity was assayed by fluorometry. Ceftriaxone treatment significantly increased SNAT1 expression and glutaminase activity in neurons in APP/PS1 mice. Similarly, VGLUT1/2 levels were increased in the presynaptic terminals of APP/PS1 mice treated with ceftriaxone. The deletion of one GLT-1 allele in APP/PS1 mice prevented the ceftriaxone-induced upregulation of SNAT1 and VGLUT1/2 expression, indicating that GLT-1 played an important role in ceftriaxone effect. Based on the role of SNAT1, glutaminase, and VGLUT1/2 in the glutamate-glutamine cycle in neurons, the present results suggested that ceftriaxone improved the production and vesicular assembly of glutamate as a neurotransmitter in presynaptic terminals by acting on GLT-1 in APP/PS1 mice.
Collapse
Affiliation(s)
- ShuJuan Fan
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, PR China
| | - Li Li
- Central Laboratory, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang 050000, PR China
| | - XiaoHui Xian
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - LiRong Liu
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, PR China
| | - JunXia Gao
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, PR China
| | - WenBin Li
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| |
Collapse
|
13
|
Resilience of network activity in preconditioned neurons exposed to 'stroke-in-a-dish' insults. Neurochem Int 2021; 146:105035. [PMID: 33798645 DOI: 10.1016/j.neuint.2021.105035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 11/22/2022]
Abstract
Exposing cultured cortical neurons to stimulatory agents - the K+ channel blocker 4-aminopyridine (4-ap), and the GABAA receptor antagonist bicuculline (bic) - for 48 h induces down-regulated synaptic scaling, and preconditions neurons to withstand subsequent otherwise lethal 'stroke-in-a-dish' insults; however, the degree to which usual neuronal function remains is unknown. As a result, multi-electrode array and patch-clamp electrophysiological techniques were employed to characterize hallmarks of spontaneous synaptic activity over a 12-day preconditioning/insult experiment. Spiking frequency increased 8-fold immediately upon 4-ap/bic treatment but declined within the 48 h treatment window to sub-baseline levels that persisted long after washout. Preconditioning resulted in key markers of network activity - spiking frequency, bursting and avalanches - being impervious to an insult. Surprisingly, preconditioning resulted in higher peak NMDA mEPSC amplitudes, resulting in a decrease in the ratio of AMPA:NMDA mEPSC currents, suggesting a relative increase in synaptic NMDA receptors. An investigation of a broad mRNA panel of excitatory and inhibitory signaling mediators indicated preconditioning rapidly up-regulated GABA synthesis (GAD67) and BDNF, followed by up-regulation of neuronal activity-regulated pentraxin and down-regulation of presynaptic glutamate release (VGLUT1). Preconditioning also enhanced surface expression of GLT-1, which persisted following an insult. Overall, preconditioning resulted in a reduced spiking frequency which was impervious to subsequent exposure to 'stroke-in-a-dish' insults, a phenotype initiated predominantly by up-regulation of inhibitory neurotransmission, a lower neuronal postsynaptic AMPA: NMDA receptor ratio, and trafficking of GLT-1 to astrocyte plasma membranes.
Collapse
|
14
|
Xian XH, Gao JX, Qi J, Fan SJ, Zhang M, Li WB. Activation of p38 MAPK participates in the sulbactam-induced cerebral ischemic tolerance mediated by glial glutamate transporter-1 upregulation in rats. Sci Rep 2020; 10:20601. [PMID: 33244020 PMCID: PMC7692545 DOI: 10.1038/s41598-020-77583-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Our previous studies have shown that sulbactam can play a neuroprotection role in hippocampal neurons by upregulating the expression and function of glial glutamate transporter-1 (GLT-1) during ischemic insult. Here, using rat global cerebral ischemia model, we studied in vivo the role of p38 mitogen-activated protein kinases (MAPK) in the sulbactam-induced GLT-1 upregulation and neuroprotection against ischemia. The hippocampal CA1 field was selected as observing target. The expressions of phosphorylated-p38 MAPK and GLT-1 were assayed with western blot analysis and immunohistochemistry. The condition of delayed neuronal death (DND) was assayed with neuropathological evaluation under thionin staining. It was shown that administration of sulbactam protected CA1 hippocampal neurons against ischemic insult accompanied with significantly upregulation in the expressions of phosphorylated-p38 MAPK and GLT-1. The time course analysis showed that sulbactam activated p38 MAPK before the GLT-1 upregulation in either normal or global cerebral ischemic rats. Furthermore, inhibiting p38 MAPK activation by SB203580 blocked the GLT-1 upregulation and neuroprotection induced by sulbactam. The above results suggested that p38 MAPK, at least partly, participated in the sulbactam-induced brain tolerance to ischemia mediated by GLT-1 upregulation in rats.
Collapse
Affiliation(s)
- Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Jun-Xia Gao
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Jie Qi
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Shu-Juan Fan
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China. .,Neuroscience Research Center of Hebei Medical University, Shijiazhuang, People's Republic of China.
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China. .,Neuroscience Research Center of Hebei Medical University, Shijiazhuang, People's Republic of China.
| |
Collapse
|
15
|
Belov Kirdajova D, Kriska J, Tureckova J, Anderova M. Ischemia-Triggered Glutamate Excitotoxicity From the Perspective of Glial Cells. Front Cell Neurosci 2020; 14:51. [PMID: 32265656 PMCID: PMC7098326 DOI: 10.3389/fncel.2020.00051] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/21/2020] [Indexed: 12/21/2022] Open
Abstract
A plethora of neurological disorders shares a final common deadly pathway known as excitotoxicity. Among these disorders, ischemic injury is a prominent cause of death and disability worldwide. Brain ischemia stems from cardiac arrest or stroke, both responsible for insufficient blood supply to the brain parenchyma. Glucose and oxygen deficiency disrupts oxidative phosphorylation, which results in energy depletion and ionic imbalance, followed by cell membrane depolarization, calcium (Ca2+) overload, and extracellular accumulation of excitatory amino acid glutamate. If tight physiological regulation fails to clear the surplus of this neurotransmitter, subsequent prolonged activation of glutamate receptors forms a vicious circle between elevated concentrations of intracellular Ca2+ ions and aberrant glutamate release, aggravating the effect of this ischemic pathway. The activation of downstream Ca2+-dependent enzymes has a catastrophic impact on nervous tissue leading to cell death, accompanied by the formation of free radicals, edema, and inflammation. After decades of “neuron-centric” approaches, recent research has also finally shed some light on the role of glial cells in neurological diseases. It is becoming more and more evident that neurons and glia depend on each other. Neuronal cells, astrocytes, microglia, NG2 glia, and oligodendrocytes all have their roles in what is known as glutamate excitotoxicity. However, who is the main contributor to the ischemic pathway, and who is the unsuspecting victim? In this review article, we summarize the so-far-revealed roles of cells in the central nervous system, with particular attention to glial cells in ischemia-induced glutamate excitotoxicity, its origins, and consequences.
Collapse
Affiliation(s)
- Denisa Belov Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
16
|
Fan S, Xian X, Li L, Yao X, Hu Y, Zhang M, Li W. Ceftriaxone Improves Cognitive Function and Upregulates GLT-1-Related Glutamate-Glutamine Cycle in APP/PS1 Mice. J Alzheimers Dis 2019; 66:1731-1743. [PMID: 30452416 DOI: 10.3233/jad-180708] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is characterized by progressive impairment of learning, memory, and cognitive deficits. Glutamate is the major excitatory neurotransmitter in the central nervous system and plays an important role in learning, memory, and cognition. The homeostasis and reutilization of glutamate are dependent on astrocytic uptake by glutamate transporter-1 (GLT-1) and the subsequent glutamate-glutamine cycle. Increasing evidence showed impairments in GLT-1 expression and uptake activity and glutamate-glutamine cycle in AD. Ceftriaxone (Cef) has been reported to upregulate the expression and uptake of GLT-1. Therefore, the present study was undertaken to explore whether Cef can improve cognitive deficits of APP/PS1 mice in early stage of AD by upregulating GLT-1 expression, and then promoting the glutamate-glutamine cycle. It was shown that Cef treatment significantly alleviated the cognitive deficits measured by Morris water maze test and upregulated GLT-1 protein expression in the hippocampus of APP/PS1 mice. Particularly, the activity of glutamine synthetase (GS) and the protein expression of system N glutamine transporter 1 (SN1), which are the key factors involved in the glutamate-glutamine cycle, were significantly upregulated as well after the Cef treatment. Furthermore, inhibition of GLT-1 uptake activity by dihydrokainic acid, an inhibitor of GLT-1, blocked the Cef-induced improvement on the cognitive deficits, GS activity, and SN1 expression. The above results suggested that Cef could improve cognitive deficits of APP/PS1 mice in early stage of AD by upregulating the GLT-1 expression, GS activity, and SN1 expression, which would lead to stimulating the glutamate-glutamine cycle.
Collapse
Affiliation(s)
- ShuJuan Fan
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - XiaoHui Xian
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - Li Li
- Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - XiaoGuang Yao
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - YuYan Hu
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - Min Zhang
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - WenBin Li
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China.,Aging and Cognition Neuroscience Laboratory of Hebei Province, Shijiazhuang, P.R. China
| |
Collapse
|
17
|
Zhao CC, Jiang MY, Zhang LY, Hu YY, Hu ZJ, Zhang MY, Qi J, Su AC, Lou N, Xian XH, Zhang JG, Li WB, Zhang M. Peroxisome proliferator-activated receptor gamma participates in the acquisition of brain ischemic tolerance induced by ischemic preconditioning via glial glutamate transporter 1 in vivo and in vitro. J Neurochem 2019; 151:608-625. [PMID: 31314916 DOI: 10.1111/jnc.14824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/08/2019] [Accepted: 07/11/2019] [Indexed: 01/29/2023]
Abstract
Glial glutamate transporter 1 (GLT-1) plays a vital role in the induction of brain ischemic tolerance (BIT) by ischemic preconditioning (IPC). However, the mechanism still needs to be further explained. The aim of this study was to investigate whether peroxisome proliferator-activated receptor gamma (PPARγ) participates in regulating GLT-1 during the acquisition of BIT induced by IPC. Initially, cerebral IPC induced BIT and enhanced PPARγ and GLT-1 expression in the CA1 hippocampus in rats. The ratio of nuclear/cytoplasmic PPARγ was also increased. At the same time, the up-regulation of PPARγ expression in astrocytes in the CA1 hippocampus was revealed by double immunofluorescence for PPARγ and glial fibrillary acidic protein. Then, the mechanism by which PPARγ regulates GLT-1 was studied in rat cortical astrocyte-neuron cocultures. We found that IPC [45 min of oxygen glucose deprivation (OGD)] protected neuronal survival after lethal OGD (4 h of OGD), which usually leads to neuronal death. The activation of PPARγ occurred earlier than the up-regulation of GLT-1 in astrocytes after IPC, as determined by western blot and immunofluorescence. Moreover, the preadministration of the PPARγ antagonist T0070907 or PPARγ siRNA significantly attenuated GLT-1 up-regulation and the neuroprotective effects induced by IPC in vitro. Finally, the effect of the PPARγ antagonist on GLT-1 expression and BIT was verified in vivo. We observed that the preadministration of T0070907 by intracerebroventricular injection dose-dependently attenuated the up-regulation of GLT-1 and BIT induced by cerebral IPC in rats. In conclusion, PPARγ participates in regulating GLT-1 during the acquisition of BIT induced by IPC. Cover Image for this issue: doi: 10.1111/jnc.14532. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Cong-Cong Zhao
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Meng-Yang Jiang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Zhen-Jie Hu
- Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Meng-Yue Zhang
- Clinical Medicine, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jie Qi
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - A-Chou Su
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Nan Lou
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Aging and Cognition Neuroscience Laboratory of Hebei Province, Shijiazhuang, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Aging and Cognition Neuroscience Laboratory of Hebei Province, Shijiazhuang, People's Republic of China
| |
Collapse
|
18
|
Huang WY, Jiang C, Ye HB, Jiao JT, Cheng C, Huang J, Liu J, Zhang R, Shao JF. miR-124 upregulates astrocytic glutamate transporter-1 via the Akt and mTOR signaling pathway post ischemic stroke. Brain Res Bull 2019; 149:231-239. [PMID: 31004734 DOI: 10.1016/j.brainresbull.2019.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 03/29/2019] [Accepted: 04/15/2019] [Indexed: 10/27/2022]
Abstract
High-concentration glutamic acid (Glu) induced by ischemic stroke can be inhibited by glutamate transporter-1 (GLT-1), which is the main mechanism for preventing excessive extracellular glutamate accumulation in the central nervous system. Upregulation of miR-124 could reduce the infarct area and promote the recovery of neurological function after ischemic stroke. A previous study investigated whether miR-124 could regulate GLT-1 expression in normal culture conditions. However, the role of miR-124 in the regulation of GLT-1 expression and further mechanisms after ischemic stroke remain unclear. In this study, the effects of miR-124 on GLT-1 expression in astrocytes after ischemic stroke were explored using an in vitro model of ischemic stroke (oxygen-glucose deprivation/reperfusion, OGD/reperfusion). The expression of GLT-1 was significantly decreased with lower expression of miR-124 in astrocytes injured by OGD/reperfusion. When miR-124 expression was improved, the expression of GLT-1 was notably increased in astrocytes injured by OGD/reperfusion. The results revealed that GLT-1 expression in astrocytes had a relationship with miR-124 after OGD/reperfusion. However, a direct interaction could not be confirmed with a luciferase reporter assay. Further results demonstrated that an inhibitor of Akt could decrease the increased protein expression of GLT-1 induced by miR-124 mimics, and an inhibitor of mTOR could increase the reduced protein expression of GLT-1 caused by a miR-124 inhibitor in astrocytes injured by different OGD/reperfusion conditions. These results indicated that miR-124 could regulate GLT-1 expression in astrocytes after OGD/reperfusion through the Akt and mTOR pathway.
Collapse
Affiliation(s)
- Wei-Yi Huang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Chen Jiang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Han-Bin Ye
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Jian-Tong Jiao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Chao Cheng
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Jin Huang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Jin Liu
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Rui Zhang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Jun-Fei Shao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China.
| |
Collapse
|
19
|
The mechanism of GLT-1 mediating cerebral ischemic injury depends on the activation of p38 MAPK. Brain Res Bull 2019; 147:1-13. [PMID: 30731111 DOI: 10.1016/j.brainresbull.2019.01.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 01/01/2019] [Accepted: 01/29/2019] [Indexed: 12/15/2022]
Abstract
The previous studies have shown that glial glutamate transporter-1 (GLT-1) participates in cerebral ischemic injury in rats. However, the mechanism involved remains to be elucidated. This study was undertaken to investigate whether p38 MAPK was involved in regulating GLT-1 in the process. At first, it was observed that global brain ischemia for 8 min led to obvious delayed neuronal death, GLT-1 down-regulation and p-p38 MAPK up-regulation in CA1 hippocampus in rats. Then, whether p-p38 MAPK was involved in regulating GLT-1 during cerebral ischemic injury was studied in vitro. Astrocyte-neuron co-cultures exposed to oxygen and glucose deprivation (OGD) were used to mimic brain ischemia. It was observed that lethal OGD (4-h OGD) decreased GLT-1 expression and increased p-p38 MAPK expression in astrocytes. The p-p38 MAPK protein rised from 0 min to 48 h that is the end time of the observation, and the peak value was at 12 h, which was 12.45 times of the control group. Moreover, pre-administration of p38 MAPK inhibitor SB203580 or its siRNA dose-dependently increased GLT-1 expression, and meanwhile alleviated the neuronal death induced by lethal OGD. The above results indicated that p38 MAPK signaling pathway participated in regulating GLT-1 during OGD injury in vitro. Finally, back to in vivo experiment, it was found that pre-administration of SB203580 by intracerebroventricular injection dose-dependently reversed the down-regulation of GLT-1 expression and attenuated the delayed neuronal death normally induced by global brain ischemia in CA1 hippocampus in rats. Taken together, it can be concluded that the mechanism of GLT-1 mediating cerebral ischemic injury depends on the activation of p38 MAPK.
Collapse
|
20
|
Li K, Zhou H, Zhan L, Shi Z, Sun W, Liu D, Liu L, Liang D, Tan Y, Xu W, Xu E. Hypoxic Preconditioning Maintains GLT-1 Against Transient Global Cerebral Ischemia Through Upregulating Cx43 and Inhibiting c-Src. Front Mol Neurosci 2018; 11:344. [PMID: 30323740 PMCID: PMC6172853 DOI: 10.3389/fnmol.2018.00344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 09/03/2018] [Indexed: 01/06/2023] Open
Abstract
Transient global cerebral ischemia (tGCI) causes excessive release of glutamate from neurons. Astrocytic glutamate transporter-1 (GLT-1) and glutamine synthetase (GS) together play a predominant role in maintaining glutamate at normal extracellular concentrations. Though our previous studies reported the alleviation of tGCI-induced neuronal death by hypoxic preconditioning (HPC) in hippocampal Cornu Ammonis 1 (CA1) of adult rats, the underlying mechanism has not yet been fully elaborated. In this study, we aimed to investigate the roles of GLT-1 and GS in the neuroprotection mediated by HPC against tGCI and to ascertain whether these roles can be regulated by connexin 43 (Cx43) and cellular-Src (c-Src) activity. We found that HPC decreased the level of extracellular glutamate in CA1 after tGCI via maintenance of GLT-1 expression and GS activity. Inhibition of GLT-1 expression with dihydrokainate (DHK) or inhibition of GS activity with methionine sulfoximine (MSO) abolished the neuroprotection induced by HPC. Also, HPC markedly upregulated Cx43 and inhibited p-c-Src expression in CA1 after tGCI, whereas inhibition of Cx43 with Gap26 dramatically reversed this effect. Furthermore, inhibition of p-c-Src with 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo (3, 4-d) pyrimidine (PP2) decreased c-Src activity, increased protein levels of GLT-1 and Cx43, enhanced GS activity, and thus reduced extracellular glutamate level in CA1 after tGCI. Collectively, our data demonstrated that reduced extracellular glutamate induced by HPC against tGCI through preventing the reduction of GLT-1 expression and maintaining GS activity in hippocampal CA1, which was mediated by upregulating Cx43 expression and inhibiting c-Src activity.
Collapse
Affiliation(s)
- Kongping Li
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Huarong Zhou
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Lixuan Zhan
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Zhe Shi
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Weiwen Sun
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Dandan Liu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Liu Liu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Donghai Liang
- Department of Environmental Health Sciences, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Yafu Tan
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China.,Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wensheng Xu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - En Xu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| |
Collapse
|
21
|
Huang YJ, Yuan YJ, Liu YX, Zhang MY, Zhang JG, Wang TC, Zhang LN, Hu YY, Li L, Xian XH, Qi J, Zhang M. Nitric Oxide Participates in the Brain Ischemic Tolerance Induced by Intermittent Hypobaric Hypoxia in the Hippocampal CA1 Subfield in Rats. Neurochem Res 2018; 43:1779-1790. [PMID: 29995175 DOI: 10.1007/s11064-018-2593-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/23/2018] [Accepted: 07/05/2018] [Indexed: 12/30/2022]
Abstract
Previous studies have shown that intermittent hypobaric hypoxia (IH) preconditioning protected neurons survival from brain ischemia. However, the mechanism remains to be elucidated. The present study explored the role of nitric oxide (NO) in the process by measuring the expression of NO synthase (NOS) and NO levels. Male Wistar rats (100) were randomly assigned into four groups: sham group, IH + sham group, ischemia group and IH + ischemia group. Rats for IH preconditioning were exposed to hypobaric hypoxia mimicking 5000 m high-altitude (PB = 404 mmHg, PO2 = 84 mmHg) 6 h/day, once daily for 28 days. Global brain ischemia was established by four-vessel occlusion that has been created by Pulsinelli. Rats were sacrificed at 7th day after the ischemia for neuropathological evaluation by thionin stain. In addition, the expression of neuronal NOS (nNOS), inducible NOS (iNOS), and NO content in the hippocampal CA1 subfield were measured at 2nd day and 7th day after the ischemia. Results revealed that global brain ischemia engendered delayed neuronal death (DND), both nNOS and iNOS expression up-regulated, and NO content increased in the hippocampal CA1 subfield. IH preconditioning reduced neuronal injury induced by the ischemia, and prevented the up-regulation of NOS expression and NO production. In addition, L-NAME + ischemia group was designed to detect whether depressing NO production could alleviate the DND. Pre-administration of L-NAME alleviated DND induced by the ischemia. These results suggest that IH preconditioning plays a protective role by inhibiting the over expression of NOS and NO content after brain ischemia.
Collapse
Affiliation(s)
- Ya-Jie Huang
- Undergraduate of Clinical Medicine, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Yu-Jia Yuan
- Undergraduate of Clinical Medicine, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Yi-Xian Liu
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Meng-Yue Zhang
- Undergraduate of Clinical Medicine, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China.
| | - Tian-Ci Wang
- Undergraduate of Clinical Medicine, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Li-Nan Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Li Li
- Department of Science and Technology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Jie Qi
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China. .,Aging and Cognition Neuroscience Laboratory of Hebei Province, Shijiazhuang, 050017, People's Republic of China.
| |
Collapse
|
22
|
Wang D, Wang X. GLT-1 mediates exercise-induced fatigue through modulation of glutamate and lactate in rats. Neuropathology 2018; 38:237-246. [DOI: 10.1111/neup.12465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Dongmei Wang
- Physical Education and Sports College; Beijing Normal University; Beijing China
- College of Sports Medicine and Rehabilitation; Taishan Medical University; Tai’an China
| | - Xingtong Wang
- College of Sports Medicine and Rehabilitation; Taishan Medical University; Tai’an China
| |
Collapse
|
23
|
Koizumi S, Hirayama Y, Morizawa YM. New roles of reactive astrocytes in the brain; an organizer of cerebral ischemia. Neurochem Int 2018; 119:107-114. [PMID: 29360494 DOI: 10.1016/j.neuint.2018.01.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 12/18/2017] [Accepted: 01/16/2018] [Indexed: 01/16/2023]
Abstract
The brain consists of neurons and much higher number of glial cells. They communicate each other, by which they control brain functions. The brain is highly vulnerable to several insults such as ischemia, but has a self-protective and self-repairing mechanisms against these. Ischemic tolerance or preconditioning is an endogenous neuroprotective phenomenon, where a mild non-lethal ischemic episode can induce resistance to a subsequent severe ischemic injury in the brain. Because of its neuroprotective effects against cerebral ischemia or stroke, ischemic tolerance has been widely studied. However, almost all studies have been performed from the viewpoint of neurons. Glial cells are structurally in close association with synapses. Recent studies have uncovered the active roles of astrocytes in modulating synaptic connectivity, such as synapse formation, elimination and maturation, during development or pathology. However, glia-mediated ischemic tolerance and/or neuronal repairing have received only limited attention. We and others have demonstrated that glial cells, especially astrocytes, play a pivotal role in regulation of induction of ischemic tolerance as well as repairing/remodeling of neuronal networks by phagocytosis. Here, we review our current understanding of (1) glial-mediated ischemic tolerance and (2) glia-mediated repairing/remodeling of the penumbra neuronal networks, and highlight their mechanisms as well as their potential benefits, problems, and therapeutic application.
Collapse
Affiliation(s)
- Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan.
| | - Yuri Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Yosuke M Morizawa
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| |
Collapse
|
24
|
Astrocytes and ischemic tolerance. Neurosci Res 2017; 126:53-59. [PMID: 29225139 DOI: 10.1016/j.neures.2017.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 11/22/2022]
Abstract
A mild non-lethal ischemic episode can induce resistance to a subsequent severe ischemic injury in the brain. This phenomenon is termed ischemic tolerance or ischemic preconditioning, and is an endogenous mechanism that can provide robust neuroprotection. Because of its neuroprotective effects against cerebral ischemia or stroke, ischemic tolerance has been widely studied. However, almost all studies have been performed from the viewpoint of neurons. Accumulating evidence suggests that glial cells have various roles in regulation of brain function, including modulation of synaptic transmission, neuronal excitation, and neuronal structure. In addition, astrocytes are closely related to homeostasis, stability of brain function, and protection of neurons. However, glial cells have received only limited attention with regard to ischemic tolerance. Cross-ischemic preconditioning is a phenomenon whereby non-ischemic preconditioning such as mechanical, thermal, and chemical treatment can induce ischemic tolerance. Of these, chemical treatments that affect the immune system can strongly induce ischemic tolerance, suggesting that glial cells may have important roles in this process. Indeed, we and others have demonstrated that glial cells, especially astrocytes, play a pivotal role in the induction of ischemic tolerance. This glial-mediated ischemic tolerance provides a robust and long-lasting neuroprotection against ischemic injury. In this review, we discuss the mechanisms underlying glial-mediated ischemic tolerance, as well as its potential benefits, problems, and therapeutic application.
Collapse
|
25
|
Krzyżanowska W, Pomierny B, Bystrowska B, Pomierny-Chamioło L, Filip M, Budziszewska B, Pera J. Ceftriaxone- and N-acetylcysteine-induced brain tolerance to ischemia: Influence on glutamate levels in focal cerebral ischemia. PLoS One 2017; 12:e0186243. [PMID: 29045497 PMCID: PMC5646803 DOI: 10.1371/journal.pone.0186243] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/27/2017] [Indexed: 01/22/2023] Open
Abstract
One of the major players in the pathophysiology of cerebral ischemia is disrupted homeostasis of glutamatergic neurotransmission, resulting in elevated extracellular glutamate (Glu) concentrations and excitotoxicity-related cell death. In the brain, Glu concentrations are regulated by Glu transporters, including Glu transporter-1 (GLT-1) and cystine/Glu antiporter (system xc-). Modulation of these transporters by administration of ceftriaxone (CEF, 200 mg/kg, i.p.) or N-acetylcysteine (NAC, 150 mg/kg, i.p.) for 5 days before focal cerebral ischemia may induce brain tolerance to ischemia by significantly limiting stroke-related damage and normalizing Glu concentrations. In the present study, focal cerebral ischemia was induced by 90-minute middle cerebral artery occlusion (MCAO). We compared the effects of CEF and NAC pretreatment on Glu concentrations in extracellular fluid and cellular-specific expression of GLT-1 and xCT with the effects of two reference preconditioning methods, namely, ischemic preconditioning and chemical preconditioning in rats. Both CEF and NAC significantly reduced Glu levels in the frontal cortex and hippocampus during focal cerebral ischemia, and this decrease was comparable with the Glu level achieved with the reference preconditioning strategies. The results of immunofluorescence staining of GLT-1 and xCT on astrocytes, neurons and microglia accounted for the observed changes in extracellular Glu levels to a certain extent. Briefly, after MCAO, the expression of GLT-1 on astrocytes decreased, but pretreatment with CEF seemed to prevent this downregulation. In addition, every intervention used in this study seemed to reduce xCT expression on astrocytes and neurons. The results of this study indicate that modulation of Glu transporter expression may restore Glu homeostasis. Moreover, our results suggest that CEF and NAC may induce brain tolerance to ischemia by influencing GLT-1 and system xc- expression levels. These transporters are presumably good targets for the development of novel therapies for brain ischemia.
Collapse
Affiliation(s)
- Weronika Krzyżanowska
- Department of Biochemical Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Bartosz Pomierny
- Department of Biochemical Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
- * E-mail:
| | - Beata Bystrowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Lucyna Pomierny-Chamioło
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Małgorzata Filip
- Institute of Pharmacology, Polish Academy of Sciences, Laboratory of Drug Addiction Pharmacology, Kraków, Poland
| | - Bogusława Budziszewska
- Department of Biochemical Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Joanna Pera
- Department of Neurology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
26
|
Lee KE, Cho KO, Choi YS, Kim SY. The neuroprotective mechanism of ampicillin in a mouse model of transient forebrain ischemia. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:185-92. [PMID: 26937215 PMCID: PMC4770109 DOI: 10.4196/kjpp.2016.20.2.185] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 01/16/2016] [Accepted: 01/23/2016] [Indexed: 11/22/2022]
Abstract
Ampicillin, a β-lactam antibiotic, dose-dependently protects neurons against ischemic brain injury. The present study was performed to investigate the neuroprotective mechanism of ampicillin in a mouse model of transient global forebrain ischemia. Male C57BL/6 mice were anesthetized with halothane and subjected to bilateral common carotid artery occlusion for 40 min. Before transient forebrain ischemia, ampicillin (200 mg/kg, intraperitoneally [i.p.]) or penicillin G (6,000 U/kg or 20,000 U/kg, i.p.) was administered daily for 5 days. The pretreatment with ampicillin but not with penicillin G signifi cantly attenuated neuronal damage in the hippocampal CA1 subfield. Mechanistically, the increased activity of matrix metalloproteinases (MMPs) following forebrain ischemia was also attenuated by ampicillin treatment. In addition, the ampicillin treatment reversed increased immunoreactivities to glial fibrillary acidic protein and isolectin B4, markers of astrocytes and microglia, respectively. Furthermore, the ampicillin treatment significantly increased the level of glutamate transporter-1, and dihydrokainic acid (DHK, 10 mg/kg, i.p.), an inhibitor of glutamate transporter-1 (GLT-1), reversed the neuroprotective effect of ampicillin. Taken together, these data indicate that ampicillin provides neuroprotection against ischemia-reperfusion brain injury, possibly through inducing the GLT-1 protein and inhibiting the activity of MMP in the mouse hippocampus.
Collapse
Affiliation(s)
- Kyung-Eon Lee
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Kyung-Ok Cho
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Yun-Sik Choi
- Department of Pharmaceutical Science and Technology, College of Health and Medical Science, Catholic University of Daegu, Daegu 712-702, Korea
| | - Seong Yun Kim
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| |
Collapse
|
27
|
N-Acetylcysteine and Ceftriaxone as Preconditioning Strategies in Focal Brain Ischemia: Influence on Glutamate Transporters Expression. Neurotox Res 2016; 29:539-50. [PMID: 26861954 PMCID: PMC4820483 DOI: 10.1007/s12640-016-9602-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 12/15/2015] [Accepted: 01/27/2016] [Indexed: 12/19/2022]
Abstract
Glutamate (Glu) plays a key role in excitotoxicity-related injury in cerebral ischemia. In the brain, Glu homeostasis depends on Glu transporters, including the excitatory amino acid transporters and the cysteine/Glu antiporter (xc-). We hypothesized that drugs acting on Glu transporters, such as ceftriaxone (CEF, 200 mg/kg, i.p.) and N-acetylcysteine (NAC, 150 mg/kg, i.p.), administered repeatedly for 5 days before focal cerebral ischemia in rats and induced by a 90-min middle cerebral artery occlusion (MCAO), may induce brain tolerance to ischemia. We compared the effects of these drugs on brain infarct volume, neurological deficits and the mRNA and protein expression of the Glu transporter-1 (GLT-1) and xc- with the effects of ischemic preconditioning and chemical preconditioning using 3-nitropropionic acid. Administration of CEF and NAC significantly reduced infarct size and neurological deficits caused by a 90-min MCAO. These beneficial effects were accompanied by changes in GLT-1 expression caused by a 90-min MCAO at both the mRNA and protein levels in the frontal cortex, hippocampus, and dorsal striatum. Thus, the results of this study suggest that the regulation of GLT-1 and xc- plays a role in the development of cerebral tolerance to ischemia and that this regulation may be a novel approach in the therapy of brain ischemia.
Collapse
|
28
|
p38 MAPK Participates in the Mediation of GLT-1 Up-regulation During the Induction of Brain Ischemic Tolerance by Cerebral Ischemic Preconditioning. Mol Neurobiol 2016; 54:58-71. [DOI: 10.1007/s12035-015-9652-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/17/2015] [Indexed: 10/22/2022]
|
29
|
Yakovlev AA, Gulyaeva NV. Possible role of proteases in preconditioning of brain cells to pathological conditions. BIOCHEMISTRY (MOSCOW) 2015; 80:163-71. [PMID: 25756531 DOI: 10.1134/s0006297915020030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Preconditioning (PC) is one of the most effective strategies to reduce the severity of cell damage, in particular of nervous tissue cells. Although PC mechanisms are studied insufficiently, it is clear that proteases are involved in them, but their role has yet been not studied in detail. In this work, some mechanisms of a potential recruiting of proteases in PC are considered. Our attention is mainly focused on the protease families of caspases and cathepsins and on protease receptors. We present evidence that just these proteins are involved in the PC of brain cells. A hypothesis is proposed that secreted cathepsin B is involved in the realization of PC through activation of PAR2 receptor.
Collapse
Affiliation(s)
- A A Yakovlev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia.
| | | |
Collapse
|
30
|
Chen HJ, Shen YC, Shiao YJ, Liou KT, Hsu WH, Hsieh PH, Lee CY, Chen YR, Lin YL. Multiplex Brain Proteomic Analysis Revealed the Molecular Therapeutic Effects of Buyang Huanwu Decoction on Cerebral Ischemic Stroke Mice. PLoS One 2015; 10:e0140823. [PMID: 26492191 PMCID: PMC4619651 DOI: 10.1371/journal.pone.0140823] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 09/29/2015] [Indexed: 11/18/2022] Open
Abstract
Stroke is the second-leading cause of death worldwide, and tissue plasminogen activator (TPA) is the only drug used for a limited group of stroke patients in the acute phase. Buyang Huanwu Decoction (BHD), a traditional Chinese medicine prescription, has long been used for improving neurological functional recovery in stroke. In this study, we characterized the therapeutic effect of TPA and BHD in a cerebral ischemia/reperfusion (CIR) injury mouse model using multiplex proteomics approach. After the iTRAQ-based proteomics analysis, 1310 proteins were identified from the mouse brain with <1% false discovery rate. Among them, 877 quantitative proteins, 10.26% (90/877), 1.71% (15/877), and 2.62% (23/877) of the proteins was significantly changed in the CIR, BHD treatment, and TPA treatment, respectively. Functional categorization analysis showed that BHD treatment preserved the integrity of the blood–brain barrier (BBB) (Alb, Fga, and Trf), suppressed excitotoxicity (Grm5, Gnai, and Gdi), and enhanced energy metabolism (Bdh), thereby revealing its multiple effects on ischemic stroke mice. Moreover, the neurogenesis marker doublecortin was upregulated, and the activity of glycogen synthase kinase 3 (GSK-3) and Tau was inhibited, which represented the neuroprotective effects. However, TPA treatment deteriorated BBB breakdown. This study highlights the potential of BHD in clinical applications for ischemic stroke.
Collapse
Affiliation(s)
- Hong-Jhang Chen
- National Research Institute of Chinese Medicine, Taipei, Taiwan
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yuh-Chiang Shen
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Young-Ji Shiao
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Kuo-Tong Liou
- Department of Chinese Martial Arts and Graduate Institute of Sport Coaching Science, Chinese Culture University, Taipei, Taiwan
| | - Wei-Hsiang Hsu
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Pei-Hsuan Hsieh
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Chi-Ying Lee
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Yet-Ran Chen
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
- * E-mail: (YLL); (YRC)
| | - Yun-Lian Lin
- National Research Institute of Chinese Medicine, Taipei, Taiwan
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
- * E-mail: (YLL); (YRC)
| |
Collapse
|
31
|
Guo Z, Zhang L, Wu Y, Li M, Yang X, He Z, Wu Z, Hu Y, Jia J. The role of glutamate transporter-1 in the acquisition of brain ischaemic tolerance in rats induced by electro-acupuncture pre-treatment. Brain Inj 2015; 29:396-402. [DOI: 10.3109/02699052.2014.896944] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Gao C, Zhu W, Tian L, Zhang J, Li Z. MCT4-mediated expression of EAAT1 is involved in the resistance to hypoxia injury in astrocyte-neuron co-cultures. Neurochem Res 2015; 40:818-28. [PMID: 25645447 DOI: 10.1007/s11064-015-1532-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/26/2015] [Accepted: 01/29/2015] [Indexed: 12/20/2022]
Abstract
Hypoxic stressors contribute to neuronal death in many brain diseases. Astrocyte processes surround most neurons and are therefore anatomically well-positioned to shield them from hypoxic injury. Excitatory amino acid transporters (EAATs), represent the sole mechanism of active reuptake of glutamate into the astrocytes and neurons and are essential to dampen neuronal excitation following glutamate release at synapses. Glutamate clearance impairment from any factors is bound to result in an increase in hypoxic neuronal injury. The brain energy metabolism under hypoxic conditions depends on monocarboxylate transporters (MCTs) that are expressed by neurons and glia. Previous co-immunoprecipitation experiments revealed that MCT4 directly modulate EAAT1 in astrocytes. The reduction in both surface proteins may act synergistically to induce neuronal hyperexcitability and excitotoxicity. Therefore we hypothesized that astrocytes would respond to hypoxic conditions by enhancing their expression of MCT4 and EAAT1, which, in turn, would enable them to better support neurons to survive lethal hypoxia injury. An oxygen deprivation (OD) protocol was used in primary cultures of neurons, astrocytes, and astrocytes-neurons derived from rat hippocampus, with or without MCT4-targeted short hairpin RNA (shRNA) transfection. Cell survival, expression of MCT4, EAAT1, glial fibrillary acidic protein and neuronal nuclear antigen were evaluated. OD resulted in significant cell death in neuronal cultures and up-regulation of MCT4, EAAT1 expression respectively in primary cell cultures, but no injury in neuron-astrocyte co-cultures and astrocyte cultures. However, neuronal cell death in co-cultures was increased exposure to shRNA-MCT4 prior to OD. These findings demonstrate that the MCT4-mediated expression of EAAT1 is involved in the resistance to hypoxia injury in astrocyte-neuron co-cultures.
Collapse
Affiliation(s)
- Chen Gao
- Department of Neurosurgery, AnNing Branch Hospital, Lanzhou General Hospital of Lanzhou Military Command, Lanzhou, 730070, Gansu Province, China
| | | | | | | | | |
Collapse
|
33
|
Hu YY, Xu J, Zhang M, Wang D, Li L, Li WB. Ceftriaxone modulates uptake activity of glial glutamate transporter-1 against global brain ischemia in rats. J Neurochem 2014; 132:194-205. [PMID: 25270764 DOI: 10.1111/jnc.12958] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/23/2014] [Accepted: 09/24/2014] [Indexed: 12/18/2022]
Abstract
Ceftriaxone(Cef) selectively increases the expression of glial glutamate transporter-1 (GLT-1), which was thought to be neuroprotective in some circumstances. However, the effect of Cef on glutamate uptake of GLT-1 was mostly assayed using in vitro studies such as primary neuron/astrocyte cultures or brain slices. In addition, the effect of Cef on neurons in different ischemic models was still discrepant. Therefore, this study was undertaken to observe the effect of Cef on neurons in global brain ischemia in rats, and especially to provide direct evidence of the up-regulation of GLT-1 uptake for glutamate contributing to the neuronal protection of Cef against brain ischemia. Neuropathological evaluation indicated that administration of Cef, especially pre-treatment protocols, significantly prevented delayed neuronal death in hippocampal CA1 subregion normally induced by global brain ischemia. Simultaneously, pre-administration of Cef significantly up-regulated the expression of GLT-1. Particularly, GLT-1 uptake assay with (3) H-glutamate in living cells from adult rats showed that up-regulation in glutamate uptake accompanied up-regulated GLT-1 expression. Inhibition of GLT-1 by antisense oligodeoxynucleotides or dihydrokainate significantly inhibited the Cef-induced up-regulation in GLT-1 uptake and the neuroprotective effect against global ischemia. Thus, we may conclude that Cef protects neurons against global brain ischemia via up-regulation of the expression and glutamate uptake of GLT-1. Glutamate uptake by glial glutamate transporter-1 (GLT-1) is the principal way to regulate extracellular glutamate homeostasis in central nervous system. Over-accumulation of glutamate results in excitotoxicity and injures neurons after cerebral ischemia. Ceftriaxone up-regulates GLT-1 expression and uptake of glutamate, diminishes the excitotoxicity of glutamate and then protects neurons against global brain ischemia.
Collapse
Affiliation(s)
- Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | | | | | | | | | | |
Collapse
|
34
|
Anuncibay-Soto B, Pérez-Rodríguez D, Llorente IL, Regueiro-Purriños M, Gonzalo-Orden JM, Fernández-López A. Age-dependent modifications in vascular adhesion molecules and apoptosis after 48-h reperfusion in a rat global cerebral ischemia model. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9703. [PMID: 25182537 PMCID: PMC4453934 DOI: 10.1007/s11357-014-9703-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/04/2014] [Indexed: 06/03/2023]
Abstract
Stroke is one of the leading causes of death and permanent disability in the elderly. However, most of the experimental studies on stroke are based on young animals, and we hypothesised that age can substantially affect the stroke response. The two-vessel occlusion model of global ischemia by occluding the common carotid arteries for 15 min at 40 mmHg of blood pressure was carried out in 3- and 18-month-old male Sprague-Dawley rats. The adhesion molecules E- and P-selectin, cell adhesion molecules (CAMs), both intercellular (ICAM-1) and vascular (VCAM-1), as well as glial fibrillary acidic protein (GFAP), and cleaved caspase-3 were measured at 48 h after ischemia in the cerebral cortex and hippocampus using Western blot, qPCR and immunofluorescence techniques. Diametric expression of GFAP and a different morphological pattern of caspase-3 labelling, although no changes in the cell number, were observed in the neurons of young and old animals. Expression of E-selectin and CAMs was also modified in an age- and ischemia/reperfusion-dependent manner. The hippocampus and cerebral cortex had similar response patterns for most of the markers studied. Our data suggest that old and young animals present different time-courses of neuroinflammation and apoptosis after ischemic damage. On the other hand, these results suggest that neuroinflammation is dependent on age rather than on the different vulnerability described for the hippocampus and cerebral cortex. These differences should be taken into account in searching for therapeutic targets.
Collapse
Affiliation(s)
- Berta Anuncibay-Soto
- />Área de Biología Celular, Instituto de Biomedicina, Universidad de León, Leon, Spain
| | - Diego Pérez-Rodríguez
- />Área de Biología Celular, Instituto de Biomedicina, Universidad de León, Leon, Spain
| | - Irene L Llorente
- />Área de Biología Celular, Instituto de Biomedicina, Universidad de León, Leon, Spain
| | - Marta Regueiro-Purriños
- />Área de Medicina, Cirugía y Anatomía Veterinaria, Instituto de Biomedicina, Universidad de León, Leon, Spain
| | - José Manuel Gonzalo-Orden
- />Área de Medicina, Cirugía y Anatomía Veterinaria, Instituto de Biomedicina, Universidad de León, Leon, Spain
| | | |
Collapse
|
35
|
Cui X, Li L, Hu YY, Ren S, Zhang M, Li WB. Sulbactam Plays Neuronal Protective Effect Against Brain Ischemia via Upregulating GLT1 in Rats. Mol Neurobiol 2014; 51:1322-33. [DOI: 10.1007/s12035-014-8809-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/10/2014] [Indexed: 11/25/2022]
|
36
|
Jagadapillai R, Mellen NM, Sachleben LR, Gozal E. Ceftriaxone preserves glutamate transporters and prevents intermittent hypoxia-induced vulnerability to brain excitotoxic injury. PLoS One 2014; 9:e100230. [PMID: 25014412 PMCID: PMC4094429 DOI: 10.1371/journal.pone.0100230] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 05/24/2014] [Indexed: 12/24/2022] Open
Abstract
Hypoxia alters cellular metabolism and although the effects of sustained hypoxia (SH) have been extensively studied, less is known about chronic intermittent hypoxia (IH), commonly associated with cardiovascular morbidity and stroke. We hypothesize that impaired glutamate homeostasis after chronic IH may underlie vulnerability to stroke-induced excitotoxicity. P16 organotypic hippocampal slices, cultured for 7 days were exposed for 7 days to IH (alternating 2 min 5% O2 - 15 min 21% O2), SH (5% O2) or RA (21% O2), then 3 glutamate challenges. The first and last exposures were intended as a metabolic stimulus (200 µM glutamate, 15 min); the second emulated excitotoxicity (10 mM glutamate, 10 min). GFAP, MAP2, and EAAT1, EAAT2 glutamate transporters expression were assessed after exposure to each hypoxic protocol. Additionally, cell viability was determined at baseline and after each glutamate challenge, in presence or absence of ceftriaxone that increases glutamate transporter expression. GFAP and MAP2 decreased after 7 days IH and SH. Long-term IH but not SH decreased EAAT1 and EAAT2. Excitotoxic glutamate challenge decreased cell viability and the following 200 µM exposure further increased cell death, particularly in IH-exposed slices. Ceftriaxone prevented glutamate transporter decrease and improved cell viability after IH and excitotoxicity. We conclude that IH is more detrimental to cell survival and glutamate homeostasis than SH. These findings suggest that impaired regulation of extracellular glutamate levels is implicated in the increased brain susceptibility to excitotoxic insult after long-term IH.
Collapse
Affiliation(s)
- Rekha Jagadapillai
- Department of Pediatrics - KCHRI, University of Louisville, Louisville, Kentucky, United States of America
| | - Nicholas M Mellen
- Department of Pediatrics - KCHRI, University of Louisville, Louisville, Kentucky, United States of America; Department of Physiology and Biophysics, University of Louisville, Louisville, Kentucky, United States of America
| | - Leroy R Sachleben
- Department of Pediatrics - KCHRI, University of Louisville, Louisville, Kentucky, United States of America
| | - Evelyne Gozal
- Department of Pediatrics - KCHRI, University of Louisville, Louisville, Kentucky, United States of America; Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States of America; Department of Physiology and Biophysics, University of Louisville, Louisville, Kentucky, United States of America
| |
Collapse
|
37
|
Mifsud G, Zammit C, Muscat R, Di Giovanni G, Valentino M. Oligodendrocyte pathophysiology and treatment strategies in cerebral ischemia. CNS Neurosci Ther 2014; 20:603-12. [PMID: 24703424 DOI: 10.1111/cns.12263] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/06/2014] [Accepted: 03/07/2014] [Indexed: 12/19/2022] Open
Abstract
Oligodendrocytes (OLs), the myelin-forming cells of the central nervous system, form a functional unit with axons and play a crucial role in axonal integrity. An episode of hypoxia-ischemia causes rapid and severe damage to these particularly vulnerable cells via multiple pathways such as overactivation of glutamate and ATP receptors, oxidative stress, and disruption of mitochondrial function. The cardinal effect of OL pathology is demyelination and dysmyelination, and this has profound effects on axonal function, transport, structure, metabolism, and survival. The OL is a primary target of ischemia in adult-onset stroke and especially in periventricular leukomalacia and should be considered as a primary therapeutic target in these conditions. More emphasis is needed on therapeutic strategies that target OLs, myelin, and their receptors, as these have the potential to significantly attenuate white matter injury and to establish functional recovery of white matter after stroke. In this review, we will summarize recent progress on the role of OLs in white matter ischemic injury and the current and emerging principles that form the basis for protective strategies against OL death.
Collapse
Affiliation(s)
- Gabriella Mifsud
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | | | | | | | | |
Collapse
|
38
|
Gong J, Gong S, Zhang M, Zhang L, Hu Y, Liu Y, Li W. Cerebral ischemic preconditioning reduces glutamate excitotoxicity by up-regulating the uptake activity of GLT-1 in rats. Amino Acids 2014; 46:1537-45. [PMID: 24643365 DOI: 10.1007/s00726-014-1723-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 03/04/2014] [Indexed: 12/31/2022]
Abstract
Our previous study has shown that cerebral ischemic preconditioning (CIP) can up-regulate the expression of glial glutamate transporter-1 (GLT-1) during the induction of brain ischemic tolerance in rats. The present study was undertaken to further explore the uptake activity of GLT-1 in the process by observing the changes in the concentration of extracellular glutamate with cerebral microdialysis and high-performance liquid chromatography. The results showed that a significant pulse of glutamate concentration reached the peak value of sevenfold of the basal level after lethal ischemic insult, which was associated with delayed neuronal death in the CA1 hippocampus. When the rats were pretreated 2 days before the lethal ischemic insult with CIP which protected the pyramidal neurons against delayed neuronal death, the peak value of glutamate concentration decreased to 3.9 fold of the basal level. Furthermore, pre-administration of dihydrokainate, an inhibitor of GLT-1, prevented the protective effect of CIP on ischemia-induced CA1 cell death. At the same time, compared with the CIP + Ischemia group, the peak value of glutamate concentration significantly increased and reached sixfold of the basal level. These results indicate that CIP induced brain ischemic tolerance via up-regulating GLT-1 uptake activity for glutamate and then decreasing the excitotoxicity of glutamate.
Collapse
Affiliation(s)
- Jianxue Gong
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
39
|
Inhibition of microRNA-181 reduces forebrain ischemia-induced neuronal loss. J Cereb Blood Flow Metab 2013; 33:1976-82. [PMID: 24002437 PMCID: PMC3851907 DOI: 10.1038/jcbfm.2013.157] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 07/31/2013] [Accepted: 08/02/2013] [Indexed: 01/09/2023]
Abstract
MicroRNA (miRNA), miR-181a, is enriched in the brain, and inhibition of miR-181a reduced astrocyte death in vitro and infarct volume after stroke in vivo. This study investigated the role of miR-181a in neuronal injury in vitro and hippocampal neuronal loss in vivo after forebrain ischemia. miR-181a levels were altered by transfection with mimic or antagomir. N2a cells subjected to serum deprivation and oxidative stress showed less cell death when miR-181a was reduced and increased death when miR-181a increased; protection was associated with increased Bcl-2 protein. In contrast, transfected primary neurons did not show altered levels of cell death when miR-181a levels changed. Naive male rats and rats stereotactically infused with miR-181a antagomir or control were subjected to forebrain ischemia and cornus ammonis (CA)1 neuronal survival and protein levels were assessed. Forebrain ischemia increased miR-181a expression and decreased Bcl-2 protein in the hippocampal CA1 region. miR-181a antagomir reduced miR-181a levels, reduced CA1 neuronal loss, increased Bcl-2 protein, and significantly prevented the decrease of glutamate transporter 1. Thus, miR-181a antagomir reduced evidence of astrocyte dysfunction and increased CA1 neuronal survival. miR-181a inhibition is thus a potential target in the setting of forebrain or global cerebral ischemia as well as focal ischemia.
Collapse
|
40
|
Affiliation(s)
- Sebastian Koch
- From the Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL (S.K.); and Departments of Neurosurgery and Radiology, University of California, Los Angeles, CA (N.G.)
| | - Nestor Gonzalez
- From the Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL (S.K.); and Departments of Neurosurgery and Radiology, University of California, Los Angeles, CA (N.G.)
| |
Collapse
|
41
|
DeSilva TM, Borenstein NS, Volpe JJ, Kinney HC, Rosenberg PA. Expression of EAAT2 in neurons and protoplasmic astrocytes during human cortical development. J Comp Neurol 2013; 520:3912-32. [PMID: 22522966 DOI: 10.1002/cne.23130] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The major regulators of synaptic glutamate in the cerebral cortex are the excitatory amino acid transporters 1-3 (EAAT1-3). In this study, we determined the cellular and temporal expression of EAAT1-3 in the developing human cerebral cortex. We applied single- and double-label immunocytochemistry to normative frontal or parietal (associative) cortex samples from 14 cases ranging in age from 23 gestational weeks to 2.5 postnatal years. The most striking finding was the transient expression of EAAT2 in layer V pyramidal neuronal cell bodies up until 8 postnatal months prior to its expression in protoplasmic astrocytes at 41 postconceptional weeks onward. EAAT2 was also expressed in neurons in layer I (presumed Cajal-Retzius cells), and white matter (interstitial) neurons. This expression in neurons in the developing human cortex contrasts with findings by others of transient expression exclusively in axon tracts in the developing sheep and rodent brain. With western blotting, we found that EAAT2 was expressed as a single band until 2 postnatal months, after which it was expressed as two bands. The expression of EAAT2 in pyramidal neurons during human brain development may contribute to cortical vulnerability to excitotoxicity during the critical period for perinatal hypoxic-ischemic encephalopathy. In addition, by studying the expression of EAAT1 and EAAT2 glutamate transporters, it was possible to document the development of protoplasmic astrocytes.
Collapse
Affiliation(s)
- Tara M DeSilva
- Department of Neurology and the FM Kirby Neurobiology Center, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | |
Collapse
|
42
|
Inui T, Alessandri B, Heimann A, Nishimura F, Frauenknecht K, Sommer C, Kempski O. Neuroprotective effect of ceftriaxone on the penumbra in a rat venous ischemia model. Neuroscience 2013; 242:1-10. [PMID: 23523747 DOI: 10.1016/j.neuroscience.2013.03.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 03/07/2013] [Accepted: 03/07/2013] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Glutamate transporter-1 (GLT-1) maintains low concentrations of extracellular glutamate by removing glutamate from the extracellular space. It is controversial, however, whether upregulation of GLT-1 is neuroprotective under all ischemic/hypoxic conditions. Recently, a neuroprotective effect of preconditioning with a β-lactam antibiotic ceftriaxone (CTX) that increases expression of GLT-1 has been reported in animal models of focal ischemia. On the other hand, it is said that CTX does not play a neuroprotective role in an in vitro study. Thus, we examined the effect of CTX on ischemic injury in a rat model of two-vein occlusion (2VO). This model mimics venous ischemia during, e.g. tumor surgery, a clinical situation that is best suitable for pretreatment with CTX. METHODS CTX (100mg/kg, 200mg/kg per day) or vehicle (0.9% NaCl) was intraperitoneally injected into Wistar rats for 5days before venous ischemia (n=57). Then, animals were prepared for occlusion of two adjacent cortical veins (2VO) by photothrombosis with rose bengal that was followed by KCl-induced cortical spreading depression (CSD). Infarct volume was evaluated with hematoxylin and eosin (H&E) staining 2days after venous occlusion. [(3)H]MK-801, [(3)H]AMPA and [(3)H]Muscimol ligand binding were examined autoradiographically in additional two groups without 2VO (n=5/group). Animals were injected either with NaCl (vehicle) or CTX 200mg/kg for 5days in order to evaluate whether NMDA, AMPA and GABAA ligand binding densities were affected. RESULTS CTX pretreatment reduced infarct volume compared to vehicle pretreatment (p<0.05). The effect of CTX pretreatment was attenuated by administration of the GLT-1 inhibitor, dihydrokainate (DHK) 30min before 2VO. CTX had no effect on the number of spontaneous spreading depressions after 2VO. Analysis of quantitative receptor autoradiography showed no statistically significant difference between rats after administration with CTX compared to control rats. CONCLUSIONS Pretreatment with CTX has neuroprotective potential without effect on NMDA, AMPA and GABAA receptor density and spontaneous spreading depression. This effect can be abolished by GLT-1 inhibition, indicating that upregulation of GLT-1 is an important mechanism for neuroprotective action in penumbra-like conditions, e.g. if neurosurgeons plan to occlude cerebral veins during tumor surgery.
Collapse
Affiliation(s)
- T Inui
- Institute for Neurosurgical Pathophysiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Li SQ, Li WB, Zhang M, Wu YZ, Hu YY. The role of neuroglobin in the neuroprotection of limb ischemic preconditioning in rats. Mol Neurobiol 2012. [PMID: 23180278 DOI: 10.1007/s12035-012-8373-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Recent evidence suggests that limb ischemic preconditioning (LIP) protects neurons against cerebral ischemia-reperfusion injury. However, the mechanisms of LIP are not well understood. Neuroglobin (Ngb) is a recently discovered globin that affords protection against hypoxic/ischemic brain injury. This study was performed to investigate the role of Ngb in the neuroprotection of LIP against brain ischemia and the involvements of mitochondria in the process. The rat global brain ischemic model was used, and the CA1 hippocampus was selected as the observational target. Ngb expression was investigated by RT-PCR and Western blot. Neuropathological evaluation was performed by thionin staining. Mitochondrial membrane potential (Δψm), Na(+)-K(+)-ATPase activity, and ultrastructure were examined by flow cytometry, spectrophotometry, and transmission electron microscopy, respectively. We also used Ngb antisense oligodeoxynucleotides (AS-ODNs) and Ngb inducer hemin to inhibit or mimic the effect of LIP. We found that LIP significantly up-regulated Ngb expression and protected neurons against ischemia. Furthermore, LIP effectively improved deterioration in the Δψm, mitochondrial Na(+)-K(+)-ATPase activity, and ultrastructure induced by cerebral ischemia. These effects of LIP were inhibited partly by Ngb AS-ODNs and mimicked by hemin. It could be concluded that up-regulation of Ngb expression played an important role in the neuroprotection induced by LIP, and the Ngb-mediated neuroprotection of LIP was, at least partly, associated with mitochondria.
Collapse
Affiliation(s)
- Shu-Qin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | | | | | | | | |
Collapse
|
44
|
Abstract
Aging increases the vulnerability of aging white matter to ischemic injury. Histone deacetylase (HDAC) inhibitors preserve young adult white matter structure and function during ischemia by conserving ATP and reducing excitotoxicity. In isolated optic nerve from 12-month-old mice, deprived of oxygen and glucose, we show that pan- and Class I-specific HDAC inhibitors promote functional recovery of axons. This protection correlates with preservation of axonal mitochondria. The cellular expression of HDAC 3 in the central nervous system (CNS), and HDAC 2 in optic nerve considerably changed with age, expanding to more cytoplasmic domains from nuclear compartments, suggesting that changes in glial cell protein acetylation may confer protection to aging axons. Our results indicate that manipulation of HDAC activities in glial cells may have a universal potential for stroke therapy across age groups.
Collapse
Affiliation(s)
- Selva Baltan
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| |
Collapse
|
45
|
Yang X, He Z, Zhang Q, Wu Y, Hu Y, Wang X, Li M, Wu Z, Guo Z, Guo J, Jia J. Pre-ischemic treadmill training for prevention of ischemic brain injury via regulation of glutamate and its transporter GLT-1. Int J Mol Sci 2012; 13:9447-9459. [PMID: 22949807 PMCID: PMC3431805 DOI: 10.3390/ijms13089447] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/14/2012] [Accepted: 07/19/2012] [Indexed: 12/12/2022] Open
Abstract
Pre-ischemic treadmill training exerts cerebral protection in the prevention of cerebral ischemia by alleviating neurotoxicity induced by excessive glutamate release following ischemic stroke. However, the underlying mechanism of this process remains unclear. Cerebral ischemia-reperfusion injury was observed in a rat model after 2 weeks of pre-ischemic treadmill training. Cerebrospinal fluid was collected using the microdialysis sampling method, and the concentration of glutamate was determined every 40 min from the beginning of ischemia to 4 h after reperfusion with high-performance liquid chromatography (HPLC)-fluorescence detection. At 3, 12, 24, and 48 h after ischemia, the expression of the glutamate transporter-1 (GLT-1) protein in brain tissues was determined by Western blot respectively. The effect of pre-ischemic treadmill training on glutamate concentration and GLT-1 expression after cerebral ischemia in rats along with changes in neurobehavioral score and cerebral infarct volume after 24 h ischemia yields critical information necessary to understand the protection mechanism exhibited by pre-ischemic treadmill training. The results demonstrated that pre-ischemic treadmill training up-regulates GLT-1 expression, decreases extracellular glutamate concentration, reduces cerebral infarct volume, and improves neurobehavioral score. Pre-ischemic treadmill training is likely to induce neuroprotection after cerebral ischemia by regulating GLT-1 expression, which results in re-uptake of excessive glutamate.
Collapse
Affiliation(s)
- Xiaojiao Yang
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.Y.); (Z.H.); (Q.Z.); (Y.W.); (Y.H.); (M.L.); (Z.W.); (Z.G.)
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Zhijie He
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.Y.); (Z.H.); (Q.Z.); (Y.W.); (Y.H.); (M.L.); (Z.W.); (Z.G.)
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Qi Zhang
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.Y.); (Z.H.); (Q.Z.); (Y.W.); (Y.H.); (M.L.); (Z.W.); (Z.G.)
| | - Yi Wu
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.Y.); (Z.H.); (Q.Z.); (Y.W.); (Y.H.); (M.L.); (Z.W.); (Z.G.)
- Department of Sports Medicine and Rehabilitation, Medical College of Fudan University, Shanghai 200032, China
| | - Yongshan Hu
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.Y.); (Z.H.); (Q.Z.); (Y.W.); (Y.H.); (M.L.); (Z.W.); (Z.G.)
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
- Department of Sports Medicine and Rehabilitation, Medical College of Fudan University, Shanghai 200032, China
| | - Xiaolou Wang
- The Third Teaching Hospital of Xinxiang Medical University, Xinxiang 453003, China; E-Mail:
| | - Mingfen Li
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.Y.); (Z.H.); (Q.Z.); (Y.W.); (Y.H.); (M.L.); (Z.W.); (Z.G.)
| | - Zhiyuan Wu
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.Y.); (Z.H.); (Q.Z.); (Y.W.); (Y.H.); (M.L.); (Z.W.); (Z.G.)
| | - Zhenzhen Guo
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.Y.); (Z.H.); (Q.Z.); (Y.W.); (Y.H.); (M.L.); (Z.W.); (Z.G.)
| | - Jingchun Guo
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
- Authors to whom correspondence should be addressed; E-Mails: (J.G.); (J.J.); Tel./Fax: +86-21-542-373-98 (J.G.); +86-21-528-878-20 (J.J.)
| | - Jie Jia
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.Y.); (Z.H.); (Q.Z.); (Y.W.); (Y.H.); (M.L.); (Z.W.); (Z.G.)
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
- Department of Sports Medicine and Rehabilitation, Medical College of Fudan University, Shanghai 200032, China
- Authors to whom correspondence should be addressed; E-Mails: (J.G.); (J.J.); Tel./Fax: +86-21-542-373-98 (J.G.); +86-21-528-878-20 (J.J.)
| |
Collapse
|
46
|
Tauskela JS, Aylsworth A, Hewitt M, Brunette E, Mealing GAR. Preconditioning induces tolerance by suppressing glutamate release in neuron culture ischemia models. J Neurochem 2012; 122:470-81. [DOI: 10.1111/j.1471-4159.2012.07791.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
47
|
Kitagawa K. Ischemic tolerance in the brain: endogenous adaptive machinery against ischemic stress. J Neurosci Res 2012; 90:1043-54. [PMID: 22302606 DOI: 10.1002/jnr.23005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Revised: 10/25/2011] [Accepted: 11/18/2011] [Indexed: 01/10/2023]
Abstract
Although more than 100 drugs have been examined clinically, tissue plasminogen activator remains the only drug approved for the treatment of acute ischemic stroke. Since the discovery of ischemic tolerance, it has been widely recognized that the brain possesses an endogenous protective machinery to protect against ischemic stress. Recent studies have clarified that both the upregulation of neuroprotective signaling and the downregulation of inflammatory or apoptotic pathways are involved equally in the acquisition of ischemic tolerance. The triggering stimuli for ischemic stresses are divided into hypoxic, oxidant/inflammatory, and glutamate stress. Glutamate stress, particularly the synaptic stimulation of the N-methyl-D-aspartate receptor, leads to activation of the cAMP response element-binding protein, which could subsequently induce gene expression of several neuroprotective molecules. Gene reprogramming and metabolic downregulation are intimately involved in ischemic tolerance as well as in hibernation and hypothermia. Micro-RNAs may be a key player for tuning the level of gene expression in ischemic tolerance. Future research should be performed to investigate the most effective combination for brain protection, enhancement of cell survival signaling, and inhibition of the inflammatory or apoptotic pathways.
Collapse
Affiliation(s)
- Kazuo Kitagawa
- Department of Neurology, Stroke Center, Osaka University Graduate School of Medicine, Suita, Japan.
| |
Collapse
|
48
|
Gong SJ, Chen LY, Zhang M, Gong JX, Ma YX, Zhang JM, Wang YJ, Hu YY, Sun XC, Li WB, Zhang Y. Intermittent hypobaric hypoxia preconditioning induced brain ischemic tolerance by up-regulating glial glutamate transporter-1 in rats. Neurochem Res 2011; 37:527-37. [PMID: 22076500 DOI: 10.1007/s11064-011-0639-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/14/2011] [Accepted: 10/28/2011] [Indexed: 01/14/2023]
Abstract
Several studies showed that the up-regulation of glial glutamate transporter-1 (GLT-1) participates in the acquisition of brain ischemic tolerance induced by cerebral ischemic preconditioning or ceftriaxone pretreatment in rats. To explore whether GLT-1 plays a role in the acquisition of brain ischemic tolerance induced by intermittent hypobaric hypoxia (IH) preconditioning (mimicking 5,000 m high-altitude, 6 h per day, once daily for 28 days), immunohistochemistry and western blot were used to observe the changes in the expression of GLT-1 protein in hippocampal CA1 subfield during the induction of brain ischemic tolerance by IH preconditioning, and the effect of dihydrokainate (DHK), an inhibitor of GLT-1, on the acquisition of brain ischemic tolerance in rats. The basal expression of GLT-1 protein in hippocampal CA1 subfield was significantly up-regulated by IH preconditioning, and at the same time astrocytes were activated by IH preconditioning, which appeared normal soma and aplenty slender processes. The GLT-1 expression was decreased at 7 days after 8-min global brain ischemia. When the rats were pretreated with the IH preconditioning before the global brain ischemia, the down-regulation of GLT-1 protein was prevented clearly. Neuropathological evaluation by thionin staining showed that 200 nmol DHK blocked the protective role of IH preconditioning against delayed neuronal death induced normally by 8-min global brain ischemia. Taken together, the up-regulation of GLT-1 protein participates in the acquisition of brain ischemic tolerance induced by IH preconditioning in rats.
Collapse
Affiliation(s)
- Shu-Juan Gong
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kostandy BB. The role of glutamate in neuronal ischemic injury: the role of spark in fire. Neurol Sci 2011; 33:223-37. [PMID: 22044990 DOI: 10.1007/s10072-011-0828-5] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Accepted: 10/20/2011] [Indexed: 12/21/2022]
Abstract
Although being a physiologically important excitatory neurotransmitter, glutamate plays a pivotal role in various neurological disorders including ischemic neurological diseases. Its level is increased during cerebral ischemia with excessive neurological stimulation causing the glutamate-induced neuronal toxicity, excitotoxicity, and this is considered the triggering spark in the ischemic neuronal damage. The glutamatergic stimulation will lead to rise in the intracellular sodium and calcium, and the elevated intracellular calcium will lead to mitochondrial dysfunction, activation of proteases, accumulation of reactive oxygen species and release of nitric oxide. Interruption of the cascades of glutamate-induced cell death during ischemia may provide a way to prevent, or at least reduce, the ischemic damage. Various therapeutic options are suggested interrupting the glutamatergic pathways, e.g., inhibiting the glutamate synthesis or release, increasing its clearance, blocking of its receptors or preventing the rise in intracellular calcium. Development of these strategies may provide future treatment options in the management of ischemic stroke.
Collapse
Affiliation(s)
- Botros B Kostandy
- Department of Pharmacology, Faculty of Medicine, University of Assiut, Assiut 71526, Egypt.
| |
Collapse
|
50
|
Liu YX, Zhang M, Liu LZ, Cui X, Hu YY, Li WB. The role of glutamate transporter-1a in the induction of brain ischemic tolerance in rats. Glia 2011; 60:112-24. [PMID: 21971915 DOI: 10.1002/glia.21252] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Accepted: 09/12/2011] [Indexed: 01/04/2023]
Abstract
This study was undertaken to determine the role of glutamate transporter-1a (GLT-1a), one of the splice variants of glutamate transporter-1, in the induction of brain ischemic tolerance by cerebral ischemic preconditioning (CIP). We used a rat global cerebral ischemic model and assessed changes by neuropathological evaluation, Western blot analysis, immunohistochemistry, real-time PCR, in vivo brain microdialysis, and high performance liquid chromatography. We found that CIP induced a significant upregulation of GLT-1a expression in the CA1 hippocampus in a time course corresponding to that of neuroprotection of CIP against brain ischemia. Severe brain ischemia for 8 min induced delayed downregulation of GLT-1a, an obvious increase in glutamate concentration and delayed neuronal death of the pyramidal neurons in the CA1 hippocampus. When the animals were pretreated with CIP before the severe ischemia, the above changes normally induced by the severe ischemia were effectively prevented. Importantly, such a preventive effect of CIP on these changes was significantly inhibited by intracerebroventricular administration of GLT-1a antisense oligodeoxynucleotides, which have been proven to specifically inhibit the expression of GLT-1a protein and mRNA, and had no effect on the expression of GLT-1b. In addition, the concentration of aspartate was also elevated after severe brain ischemic insult. However, CIP had no effect on the elevated aspartate concentrations. These results indicate that GLT-1a participated in the brain ischemic tolerance induced by CIP in rats.
Collapse
Affiliation(s)
- Yi-Xian Liu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | | | | | | | | | | |
Collapse
|