1
|
Lv M, Duan Z, Tan J, Liu J, Wang Q, Wang C, Zhang Z, Sun X, Liu R, Cui Y. PHGDH-mediated serine synthesis in astrocytes supports neuroinflammation by sustaining NADH level to promote histone acetylation. Cell Death Dis 2025; 16:397. [PMID: 40383841 PMCID: PMC12086227 DOI: 10.1038/s41419-025-07732-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 04/26/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Neuroinflammation contributes to the loss of dopamine neurons and motor dysfunctions in Parkinson's disease (PD). How cell metabolism regulates neuroinflammation by modulating epigenetic modifications is largely unknown. In this study, we found that the expression of phosphoglycerate dehydrogenase (PHGDH) which catalyzes the first step of the de novo serine synthesis pathway was mainly expressed in astrocytes and l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) injection triggered the upregulation of PHGDH in astrocytes in substantia nigra. PHGDH inhibition or knockdown reduced proinflammatory cytokine production in primary astrocytes after LPS (lipopolysaccharide) stimulation which was not due to suppressed inflammatory signaling transduction. Mechanistically, PHGDH promotes proinflammatory cytokine transcription by sustaining nicotinamide adenine dinucleotide (NADH) accumulation to facilitate histone acetylation of cytokine promoters. Moreover, PHGDH inhibition-induced inflammatory response decreased neurotoxicity in vitro and alleviated astrocytes-mediated neuroinflammation and neurotoxicity in an MPTP mice model. This study reveals the role and mechanism of PHGDH-mediated serine synthesis in promoting the inflammatory response of astrocytes which may provide a potential target for neurological diseases involving neuroinflammation.
Collapse
Affiliation(s)
- Mengfei Lv
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zhongying Duan
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Jinhua Tan
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Jiake Liu
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Qinqin Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong, China
| | - Congxiao Wang
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhaolong Zhang
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaona Sun
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Rui Liu
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Yu Cui
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China.
- Qingdao Medical College, Qingdao University, Qingdao, China.
| |
Collapse
|
2
|
Xu X, Duan Z, Zhou X, Zhao R, Xu J, Zhang Z, Lv M, Wan Q, Cui Y. SFXN1 Reduction Alleviates Cerebral Ischemia-Reperfusion Injury by Promoting Neuronal Survival and Reducing Neuroinflammation. CNS Neurosci Ther 2025; 31:e70457. [PMID: 40420406 PMCID: PMC12106369 DOI: 10.1111/cns.70457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/16/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
AIM Sideroflexin 1 (SFXN1) is an important inner mitochondrial membrane protein that regulates many physiological and pathological events. However, the role of SFXN1 in cerebral ischemia-reperfusion (I/R)-induced neuronal death remains unclear. METHODS We employed in vivo injury models of transient middle cerebral artery occlusion (tMCAO) and in vitro models of lipopolysaccharide (LPS) stimulation and oxygen-glucose deprivation/reperfusion (OGD/R) to investigate the regulatory effects of SFXN1 on neuroinflammation and brain injury. Western blotting, immunofluorescence, and real-time quantitative PCR were utilized to assess SFXN1 expression, proinflammatory signaling pathways activation, and cytokine levels in vitro. Cerebral infarction was evaluated using 2,3,5-triphenyltetrazolium chloride (TTC) staining and Nissl staining. RESULTS SFXN1 expression was upregulated following cerebral I/R injury. Both neurons and microglia exhibited increased SFXN1 expression after oxygen-glucose deprivation/reoxygenation (OGD/R) treatment. SFXN1 knockdown reduced OGD/R-induced neuronal death and alleviated cerebral I/R injury. Additionally, conditioned medium from SFXN1-knockdown microglia reduced neurotoxicity in vitro. Mechanistically, SFXN1 induced mitochondrial dysfunction and neuronal death after OGD/R in an iron-independent manner. Furthermore, SFXN1 promoted the production of proinflammatory cytokines by promoting NF-κB activation, partially through iron transport in microglia after OGD/R. CONCLUSION This study reveals the novel role of SFXN1 in exacerbating cerebral I/R injury by reducing neuronal survival through the modulation of mitochondrial function and promotion of microglia-mediated neuroinflammation via NF-κB activation.
Collapse
Affiliation(s)
- Xiangyu Xu
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| | - Zhongying Duan
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
- School of Basic MedicineQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| | - Xin Zhou
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
- School of Basic MedicineQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| | - Rui Zhao
- Department of Interventional RadiologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Jing Xu
- Shandong First Medical University & Shandong Academy of Medical SciencesJinanShandongChina
| | - Zhaolong Zhang
- Department of Interventional RadiologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Mengfei Lv
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| | - Qi Wan
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
- Faculty of Life and HealthShenzhen University of Advanced ScienceShenzhenChina
| | - Yu Cui
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
- School of Basic MedicineQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| |
Collapse
|
3
|
Liu W, Liu X, Liu M, Zhao R, Zhao Z, Xiao J, Wan D, Wan Q, Xu R. Oxyglutamate Carrier Alleviates Cerebral Ischaemia-Reperfusion Injury by Regulating Mitochondrial Function. Eur J Neurosci 2025; 61:e16659. [PMID: 39777930 DOI: 10.1111/ejn.16659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 11/09/2024] [Accepted: 12/13/2024] [Indexed: 01/30/2025]
Abstract
Mitochondrial dysfunction has been reported to participate in the pathophysiological processes of cerebral ischaemia-reperfusion injury, which include reduced energy homeostasis, increased generation of oxidative stress species (ROS) and the release of apoptotic factors. Oxyglutamate carrier (OGC) is an important carrier protein on the inner mitochondrial membrane that can transport metabolites from the cytoplasm to the mitochondria. The role of OGC in cerebral ischaemia-reperfusion injury (I/R) remains unknown. In this study, we found that the expression of OGC was significantly upregulated after cerebral ischaemia-reperfusion injury. Inhibiting OGC with phenylsuccinic acid (PSA) increased neuronal death after oxygen-glucose deprivation/reoxygenation (OGD/R) in vitro. Mechanistically, OGC was localized in mitochondria and could facilitate the transport of glutathione from the cytoplasm to the mitochondria to reduce ROS levels and increase ATP production after OGD/R. In addition, in vivo inhibition of OGC exacerbated brain infarction, and GSH supplementation alleviated brain infarction resulting from OGC inhibition. This study revealed the role of OGC in alleviating brain damage by regulating mitochondrial GSH transport to alleviate mitochondrial function after cerebral ischaemia-reperfusion injury, which may provide a target for alleviating ischaemic brain injury.
Collapse
Affiliation(s)
- Wenhao Liu
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xin Liu
- Department of Interventional Radiology, The People's Hospital of Rizhao, Rizhao, China
| | - Min Liu
- Department of Interventional Radiology, The People's Hospital of Rizhao, Rizhao, China
| | - Rui Zhao
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zhiyuan Zhao
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Jingrui Xiao
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Dongdong Wan
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Qi Wan
- Qingdao Medical College, Qingdao University, Qingdao, China
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
| | - Rui Xu
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Kong X, Lyu W, Lin X, Feng H, Xu L, Li C, Sun X, Lin C, Li J, Wei P. Transcranial direct current stimulation enhances the protective effect of isoflurane preconditioning on cerebral ischemia/reperfusion injury: A new mechanism associated with the nuclear protein Akirin2. CNS Neurosci Ther 2024; 30:e70033. [PMID: 39267282 PMCID: PMC11393012 DOI: 10.1111/cns.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 09/17/2024] Open
Abstract
AIMS Ischemic stroke is a major cause of disability and mortality worldwide. Transcranial direct current stimulation (tDCS) and isoflurane (ISO) preconditioning exhibit neuroprotective properties. However, it remains unclear whether tDCS enhances the protective effect of ISO preconditioning on ischemic stroke, and the underlying mechanisms are yet to be clarified. METHOD A model of middle cerebral artery occlusion (MCAO), a rat ischemia-reperfusion (I/R) injury model, and an in vitro oxygen-glucose deprivation/re-oxygenation (O/R) model of ischemic injury were developed. ISO preconditioning and tDCS were administered daily for 7 days before MCAO modeling. Triphenyltetrazolium chloride staining, modified neurological severity score, and hanging-wire test were conducted to assess infarct volume and neurological outcomes. Untargeted metabolomic experiments, adeno-associated virus, lentiviral vectors, and small interfering RNA techniques were used to explore the underlying mechanisms. RESULTS tDCS/DCS enhanced the protective effects of ISO pretreatment on I/R injury-induced brain damage. This was evidenced by reduced infarct volume and improved neurological outcomes in rats with MCAO, as well as decreased cortical neuronal death after O/R injury. Untargeted metabolomic experiments identified oxidative phosphorylation (OXPHOS) as a critical pathological process for ISO-mediated neuroprotection from I/R injury. The combination of tDCS/DCS with ISO preconditioning significantly inhibited I/R injury-induced OXPHOS. Mechanistically, Akirin2, a small nuclear protein that regulates cell proliferation and differentiation, was found to decrease in the cortex of rats with MCAO and in cortical primary neurons subjected to O/R injury. Akirin2 functions upstream of phosphatase and tensin homolog deleted on chromosome 10 (PTEN). tDCS/DCS was able to further upregulate Akirin2 levels and activate the Akirin2/PTEN signaling pathway in vivo and in vitro, compared with ISO pretreatment alone, thereby contributing to the improvement of cerebral I/R injury. CONCLUSION tDCS treatment enhances the neuroprotective effects of ISO preconditioning on ischemic stroke by inhibiting oxidative stress and activating Akirin2-PTEN signaling pathway, highlighting potential of combination therapy in ischemic stroke.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Wenyuan Lyu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Xiaojie Lin
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Hao Feng
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Lin Xu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Chengwei Li
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Xinyi Sun
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Chunlong Lin
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Jianjun Li
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Penghui Wei
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| |
Collapse
|
5
|
Kong X, Lyu W, Lin X, Lin C, Feng H, Xu L, Shan K, Wei P, Li J. Itaconate alleviates anesthesia/surgery-induced cognitive impairment by activating a Nrf2-dependent anti-neuroinflammation and neurogenesis via gut-brain axis. J Neuroinflammation 2024; 21:104. [PMID: 38649932 PMCID: PMC11034021 DOI: 10.1186/s12974-024-03103-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a common neurological complication of anesthesia and surgery in aging individuals. Neuroinflammation has been identified as a hallmark of POCD. However, safe and effective treatments of POCD are still lacking. Itaconate is an immunoregulatory metabolite derived from the tricarboxylic acid cycle that exerts anti-inflammatory effects by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. In this study, we investigated the effects and underlying mechanism of 4-octyl itaconate (OI), a cell-permeable itaconate derivative, on POCD in aged mice. METHODS A POCD animal model was established by performing aseptic laparotomy in 18-month-old male C57BL/6 mice under isoflurane anesthesia while maintaining spontaneous ventilation. OI was intraperitoneally injected into the mice after surgery. Primary microglia and neurons were isolated and treated to lipopolysaccharide (LPS), isoflurane, and OI. Cognitive function, neuroinflammatory responses, as well as levels of gut microbiota and their metabolites were evaluated. To determine the mechanisms underlying the therapeutic effects of OI in POCD, ML385, an antagonist of Nrf2, was administered intraperitoneally. Cognitive function, neuroinflammatory responses, endogenous neurogenesis, neuronal apoptosis, and Nrf2/extracellular signal-related kinases (ERK) signaling pathway were evaluated. RESULTS Our findings revealed that OI treatment significantly alleviated anesthesia/surgery-induced cognitive impairment, concomitant with reduced levels of the neuroinflammatory cytokines IL-1β and IL-6, as well as suppressed activation of microglia and astrocytes in the hippocampus. Similarly, OI treatment inhibited the expression of IL-1β and IL-6 in LPS and isoflurane-induced primary microglia in vitro. Intraperitoneal administration of OI led to alterations in the gut microbiota and promoted the production of microbiota-derived metabolites associated with neurogenesis. We further confirmed that OI promoted endogenous neurogenesis and inhibited neuronal apoptosis in the hippocampal dentate gyrus of aged mice. Mechanistically, we observed a decrease in Nrf2 expression in hippocampal neurons both in vitro and in vivo, which was reversed by OI treatment. We found that Nrf2 was required for OI treatment to inhibit neuroinflammation in POCD. The enhanced POCD recovery and promotion of neurogenesis triggered by OI exposure were, at least partially, mediated by the activation of the Nrf2/ERK signaling pathway. CONCLUSIONS Our findings demonstrate that OI can attenuate anesthesia/surgery-induced cognitive impairment by stabilizing the gut microbiota and activating Nrf2 signaling to restrict neuroinflammation and promote neurogenesis. Boosting endogenous itaconate or supplementation with exogenous itaconate derivatives may represent novel strategies for the treatment of POCD.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Wenyuan Lyu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Xiaojie Lin
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Chunlong Lin
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Hao Feng
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Lin Xu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Kaiyue Shan
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Penghui Wei
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China.
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China.
| | - Jianjun Li
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China.
- Laboratory of Anesthesia and Brain Function, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China.
| |
Collapse
|
6
|
Wang H, Ma W, Hu W, Li X, Shen N, Li Z, Kong X, Lin T, Gao J, Zhu T, Che F, Chen J, Wan Q. Cathodal bilateral transcranial direct-current stimulation regulates selenium to confer neuroprotection after rat cerebral ischaemia-reperfusion injury. J Physiol 2024; 602:1175-1197. [PMID: 38431908 DOI: 10.1113/jp285806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
Non-invasive transcranial direct-current stimulation (tDCS) is a safe ischaemic stroke therapy. Cathodal bilateral tDCS (BtDCS) is a modified tDCS approach established by us recently. Because selenium (Se) plays a crucial role in cerebral ischaemic injury, we investigated whether cathodal BtDCS conferred neuroprotection via regulating Se-dependent signalling in rat cerebral ischaemia-reperfusion (I/R) injury. We first showed that the levels of Se and its transport protein selenoprotein P (SEPP1) were reduced in the rat cortical penumbra following I/R, whereas cathodal BtDCS prevented the reduction of Se and SEPP1. Interestingly, direct-current stimulation (DCS) increased SEPP1 level in cultured astrocytes subjected to oxygen-glucose deprivation reoxygenation (OGD/R) but had no effect on SEPP1 level in OGD/R-insulted neurons, indicating that DCS may increase Se in ischaemic neurons by enhancing the synthesis and secretion of SEPP1 in astrocytes. We then revealed that DCS reduced the number of injured mitochondria in OGD/R-insulted neurons cocultured with astrocytes. DCS and BtDCS prevented the reduction of the mitochondrial quality-control signalling, vesicle-associated membrane protein 2 (VAMP2) and syntaxin-4 (STX4), in OGD/R-insulted neurons cocultured with astrocytes and the ischaemic brain respectively. Under the same experimental conditions, downregulation of SEPP1 blocked DCS- and BtDCS-induced upregulation of VAMP2 and STX4. Finally, we demonstrated that cathodal BtDCS increased Se to reduce infract volume following I/R. Together, the present study uncovered a molecular mechanism by which cathodal BtDCS confers neuroprotection through increasing SEPP1 in astrocytes and subsequent upregulation of SEPP1/VAMP2/STX4 signalling in ischaemic neurons after rat cerebral I/R injury. KEY POINTS: Cathodal bilateral transcranial direct-current stimulation (BtDCS) prevents the reduction of selenium (Se) and selenoprotein P in the ischaemic penumbra. Se plays a crucial role in cerebral ischaemia injury. Direct-current stimulation reduces mitochondria injury and blocks the reduction of vesicle-associated membrane protein 2 (VAMP2) and syntaxin-4 (STX4) in oxygen-glucose deprivation reoxygenation-insulted neurons following coculturing with astrocytes. Cathodal BtDCS regulates Se/VAMP2/STX4 signalling to confer neuroprotection after ischaemia.
Collapse
Affiliation(s)
- Hui Wang
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenlong Ma
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenjie Hu
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaohua Li
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Na Shen
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhuo Li
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiangyi Kong
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Tao Lin
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jingchen Gao
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Ting Zhu
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fengyuan Che
- Central Laboratory, Department of Neurology, Linyi People's Hospital, Qingdao University, Linyi, Shandong, China
| | - Juan Chen
- Department of Neurology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Qi Wan
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
- Qingdao Gui-Hong Intelligent Medical Technology Co. Ltd, Qingdao, China
| |
Collapse
|