1
|
Fu X, Li J, Yang S, Jing J, Zheng Q, Zhang T, Xu Z. Blood-brain barrier repair: potential and challenges of stem cells and exosomes in stroke treatment. Front Cell Neurosci 2025; 19:1536028. [PMID: 40260076 PMCID: PMC12009835 DOI: 10.3389/fncel.2025.1536028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/12/2025] [Indexed: 04/23/2025] Open
Abstract
Stroke is characterized with high morbidity, mortality and disability all over the world, and one of its core pathologies is blood-brain barrier (BBB) dysfunction. BBB plays a crucial physiological role in protecting brain tissues and maintaining homeostasis in central nervous system (CNS). BBB dysfunction serves as a key factor in the development of cerebral edema, inflammation, and further neurological damage in stroke patients. Currently, stem cells and their derived exosomes have shown remarkable potential in repairing the damaged BBB and improving neurological function after stroke. Stem cells repair the integrity of BBB through anti-inflammatory, antioxidant, angiogenesis and regulation of intercellular signaling mechanisms, while stem cell-derived exosomes, as natural nanocarriers, further enhance the therapeutic effect by carrying active substances such as proteins, RNAs and miRNAs. This review will present the latest research advances in stem cells and their exosomes in stroke treatment, as well as the challenges of cell source, transplantation timing, dosage, and route of administration in clinical application, aiming to discuss their mechanisms of repairing BBB integrity and potential for clinical application, and proposes future research directions. Stem cells and exosomes are expected to provide new strategies for early diagnosis and precise treatment of stroke, and promote breakthroughs in the field of stroke.
Collapse
Affiliation(s)
- Xiaochen Fu
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Jia Li
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Shoujun Yang
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiapeng Jing
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Qinzhi Zheng
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Ting Zhang
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
- Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, China
| | - Zhuo Xu
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Pérez-González AP, de Anda-Jáuregui G, Hernández-Lemus E. Differential Transcriptional Programs Reveal Modular Network Rearrangements Associated with Late-Onset Alzheimer's Disease. Int J Mol Sci 2025; 26:2361. [PMID: 40076979 PMCID: PMC11900169 DOI: 10.3390/ijms26052361] [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: 01/31/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is a complex, genetically heterogeneous disorder. The diverse phenotypes associated with AD result from interactions between genetic and environmental factors, influencing multiple biological pathways throughout disease progression. Network-based approaches offer a way to assess phenotype-specific states. In this study, we calculated key network metrics to characterize the network transcriptional structure and organization in LOAD, focusing on genes and pathways implicated in AD pathology within the dorsolateral prefrontal cortex (DLPFC). Our findings revealed disease-specific coexpression markers associated with diverse metabolic functions. Additionally, significant differences were observed at both the mesoscopic and local levels between AD and control networks, along with a restructuring of gene coexpression and biological functions into distinct transcriptional modules. These results show the molecular reorganization of the transcriptional program occurring in LOAD, highlighting specific adaptations that may contribute to or result from cellular responses to pathological stressors. Our findings may support the development of a unified model for the causal mechanisms of AD, suggesting that its diverse manifestations arise from multiple pathways working together to produce the disease's complex clinical patho-phenotype.
Collapse
Affiliation(s)
- Alejandra Paulina Pérez-González
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Programa de Doctorado en Ciencias Biomédicas, Unidad de Posgrado Edificio B Primer Piso, Ciudad Universitaria, Mexico City 04510, Mexico
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
| | - Guillermo de Anda-Jáuregui
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Investigadores por M’exico, Conahcyt, Mexico City 03940, Mexico
| | - Enrique Hernández-Lemus
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
3
|
Yuan X, Li W, Yan Q, Ou Y, Long Q, Zhang P. Biomarkers of mature neuronal differentiation and related diseases. Future Sci OA 2024; 10:2410146. [PMID: 39429212 PMCID: PMC11497955 DOI: 10.1080/20565623.2024.2410146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/16/2024] [Indexed: 10/22/2024] Open
Abstract
The nervous system regulates perception, cognition and behavioral responses by serving as the body's primary communication system for receiving, regulating and transmitting information. Neurons are the fundamental structures and units of the nervous system. Their differentiation and maturation processes rely on the expression of specific biomarkers. Neuron-specific intracellular markers can be used to determine the degree of neuronal maturation. Neuronal cytoskeletal proteins dictate the shape and structure of neurons, while synaptic plasticity and signaling processes are intricately associated with neuronal synaptic markers. Furthermore, abnormal expression levels of biomarkers can serve as diagnostic indicators for nervous system diseases. This article reviews the markers of mature neuronal differentiation and their relationship with nervous system diseases.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Wen Li
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Ya Ou
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Qingxi Long
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| |
Collapse
|
4
|
Wang XP, Yan D, Jin XP, Zhang WY, Shi T, Wang X, Song W, Xiong X, Guo D, Chen S. The role of amino acid metabolism alterations in acute ischemic stroke: From mechanism to application. Pharmacol Res 2024; 207:107313. [PMID: 39025169 DOI: 10.1016/j.phrs.2024.107313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Acute ischemic stroke (AIS) is the most prevalent type of stroke, and due to its high incidence, disability rate, and mortality rate, it imposes a significant burden on the health care system. Amino acids constitute one of the most crucial metabolic products within the human body, and alterations in their metabolic pathways have been identified in the microenvironment of AIS, thereby influencing the pathogenesis, severity, and prognosis of AIS. The amino acid metabolism characteristics in AIS are complex. On one hand, the dynamic progression of AIS continuously reshapes the amino acid metabolism pattern. Conversely, changes in the amino acid metabolism pattern also exert a double-edged effect on AIS. This interaction is bidirectional, dynamic, heterogeneous, and dose-specific. Therefore, the distinctive metabolic reprogramming features surrounding amino acids during the AIS process are systematically summarized in this paper, aiming to provide potential investigative strategies for the early diagnosis, treatment approaches, and prognostic enhancement of AIS.
Collapse
Affiliation(s)
- Xiang-Ping Wang
- First People's Hospital of Linping District; Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 311100, China
| | - Dan Yan
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou 311202, China
| | - Xia-Ping Jin
- First People's Hospital of Linping District; Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 311100, China
| | - Wen-Yan Zhang
- First People's Hospital of Linping District; Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 311100, China
| | - Tao Shi
- First People's Hospital of Linping District; Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 311100, China
| | - Xiang Wang
- First People's Hospital of Linping District; Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 311100, China
| | - Wenjuan Song
- First People's Hospital of Linping District; Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 311100, China
| | - Xing Xiong
- Traditional Chinese Medical Hospital of Xiaoshan, The Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 311200, China
| | - Duancheng Guo
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Sheng Chen
- First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang Province 311200, China.
| |
Collapse
|
5
|
Bugaj AM, Kunath N, Saasen VL, Fernandez-Berrocal MS, Vankova A, Sætrom P, Bjørås M, Ye J. Dissecting gene expression networks in the developing hippocampus through the lens of NEIL3 depletion. Prog Neurobiol 2024; 235:102599. [PMID: 38522610 DOI: 10.1016/j.pneurobio.2024.102599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/09/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Gene regulation in the hippocampus is fundamental for its development, synaptic plasticity, memory formation, and adaptability. Comparisons of gene expression among different developmental stages, distinct cell types, and specific experimental conditions have identified differentially expressed genes contributing to the organization and functionality of hippocampal circuits. The NEIL3 DNA glycosylase, one of the DNA repair enzymes, plays an important role in hippocampal maturation and neuron functionality by shaping transcription. While differential gene expression (DGE) analysis has identified key genes involved, broader gene expression patterns crucial for high-order hippocampal functions remain uncharted. By utilizing the weighted gene co-expression network analysis (WGCNA), we mapped gene expression networks in immature (p8-neonatal) and mature (3 m-adult) hippocampal circuits in wild-type and NEIL3-deficient mice. Our study unveiled intricate gene network structures underlying hippocampal maturation, delineated modules of co-expressed genes, and pinpointed highly interconnected hub genes specific to the maturity of hippocampal subregions. We investigated variations within distinct gene network modules following NEIL3 depletion, uncovering NEIL3-targeted hub genes that influence module connectivity and specificity. By integrating WGCNA with DGE, we delve deeper into the NEIL3-dependent molecular intricacies of hippocampal maturation. This study provides a comprehensive systems-level analysis for assessing the potential correlation between gene connectivity and functional connectivity within the hippocampal network, thus shaping hippocampal function throughout development.
Collapse
Affiliation(s)
- Anna M Bugaj
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Nicolas Kunath
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway; Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway; Department of Neurology, University Hospital of Trondheim, Trondheim 7491, Norway
| | - Vidar Langseth Saasen
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Marion S Fernandez-Berrocal
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Ana Vankova
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Pål Sætrom
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway; Department of Microbiology, Oslo University Hospital, University of Oslo, Oslo 0424, Norway; Centre for Embryology and Healthy Development, University of Oslo, Oslo 0373, Norway.
| | - Jing Ye
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway.
| |
Collapse
|
6
|
Yan C, Liu Z, Xie W, Zhang T, Zhang J, Li G, Xu X, Ye L, Gong J. Cornuside protects against ischemic stroke in rats by suppressing the IL-17F/TRAF6/NF-κB pathway via the brain-gut axis. Exp Neurol 2024; 373:114672. [PMID: 38169196 DOI: 10.1016/j.expneurol.2023.114672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024]
Abstract
Ischemic stroke is a serious neurological disease with limited therapeutic options; thus, it is particularly important to find effective treatments. Restoration of gut microflora diversity is an important factor in the treatment of ischemic stroke, but the mechanism remains unclear. Cornuside is known for its unique anti-inflammatory and circulation-promoting effects; however, whether it can effectively treat ischemic stroke and its therapeutic mechanisms remain unknown. In this study, we used a rat middle cerebral artery occlusion-reperfusion model (MCAO/R) to mimic ischemic stroke in humans and to assess the cerebral protective effects of cornuside in rats with ischemic stroke. Using 16S rRNA sequencing and RNA sequencing, we explored the cornuside mechanism in the brain-gut axis that confers protection against ischemic stroke. In conclusion, cornuside can inhibit the IL-17F/TRAF6/NF-κB pathway by improving the dysregulation of intestinal microflora, and reduce intestinal inflammation and neuroinflammation, which treated ischemic stroke rats.
Collapse
Affiliation(s)
- Chao Yan
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Zhihao Liu
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Waner Xie
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Ting Zhang
- Key Laboratory for Genetic Hearing Disorders in Shandong, Department of human anatomy, Binzhou Medical University, Yantai 264003, PR China
| | - Jiyao Zhang
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Guodong Li
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Xiaoyan Xu
- Department of Immunology, Binzhou Medical University, Yantai 264003, PR China.
| | - Lei Ye
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 263003, Shandong, PR China.
| | - Jianwei Gong
- Department of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, PR China.
| |
Collapse
|
7
|
Zhu Y, Hui Q, Zhang Z, Fu H, Qin Y, Zhao Q, Li Q, Zhang J, Guo L, He W, Han C. Advancements in the study of synaptic plasticity and mitochondrial autophagy relationship. J Neurosci Res 2024; 102:e25309. [PMID: 38400573 DOI: 10.1002/jnr.25309] [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: 10/09/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Synapses serve as the points of communication between neurons, consisting primarily of three components: the presynaptic membrane, synaptic cleft, and postsynaptic membrane. They transmit signals through the release and reception of neurotransmitters. Synaptic plasticity, the ability of synapses to undergo structural and functional changes, is influenced by proteins such as growth-associated proteins, synaptic vesicle proteins, postsynaptic density proteins, and neurotrophic growth factors. Furthermore, maintaining synaptic plasticity consumes more than half of the brain's energy, with a significant portion of this energy originating from ATP generated through mitochondrial energy metabolism. Consequently, the quantity, distribution, transport, and function of mitochondria impact the stability of brain energy metabolism, thereby participating in the regulation of fundamental processes in synaptic plasticity, including neuronal differentiation, neurite outgrowth, synapse formation, and neurotransmitter release. This article provides a comprehensive overview of the proteins associated with presynaptic plasticity, postsynaptic plasticity, and common factors between the two, as well as the relationship between mitochondrial energy metabolism and synaptic plasticity.
Collapse
Affiliation(s)
- Yousong Zhu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinlong Hui
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Zheng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Hao Fu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Yali Qin
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qiong Zhao
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinqing Li
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Junlong Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Lei Guo
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Wenbin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Cheng Han
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
8
|
Cao C, Lu T, Cheng Q, Cui G, Wang Z, Li X, Li H, Gao H, Shen H, Sun Q. Restoring System xc- activity by xCT overexpression inhibited neuronal ferroptosis and improved neurological deficits after experimental subarachnoid hemorrhage. Brain Res 2023; 1820:148556. [PMID: 37648093 DOI: 10.1016/j.brainres.2023.148556] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Ferroptosis is an important therapeutic target to alleviate early brain injury (EBI) after subarachnoid hemorrhage (SAH), yet the mechanism of neuronal ferroptosis after SAH remains unclear. System xc- dysfunction is one of the key pathways to induce ferroptosis. System xc- activity is mainly regulated by the expression of xCT. This study was designed to investigate the effect of xCT expression and System xc- activity on ferroptosis and EBI in an experimental SAH model both in vitro and in vivo. METHODS SAH was induced in adult male Sprague-Dawley rats by injecting autologous blood into the prechiasmatic cistern. Primary neurons treated with oxyhemoglobin (10 µM) were used to mimic SAH in vitro. Plasmid transfection was used to induce xCT overexpression. Western blotting, immunofluorescence staining, measurement of cystine uptake, enzyme-linked immunosorbent assay, transmission electron microscopy, Nissl staining, and a series of neurobehavioral tests were conducted to explore the role of xCT and System xc- activity in ferroptosis and EBI after SAH. RESULTS We found that System xc- dysfunction induced ferroptosis and exacerbated EBI after SAH in rats. xCT deficiency after SAH resulted in System xc- dysfunction, weakened neuronal antioxidant capacity and activated neuronal ferroptosis. xCT overexpression improved neuronal antioxidant capacity and inhibited neuronal ferroptosis by restoring System xc- activity. Rats with xCT overexpression after SAH presented with attenuated brain edema and inflammation, increased neuronal survival, and ameliorated neurological deficits. CONCLUSIONS Our study revealed that restoring System xc- activity by xCT overexpression inhibited neuronal ferroptosis and EBI and improved neurological deficits after SAH.
Collapse
Affiliation(s)
- Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Intensive Care Unit, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China; Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China.
| | - Ting Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Qian Cheng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Gang Cui
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Heng Gao
- Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China.
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| |
Collapse
|
9
|
Shao J, Meng Y, Yuan K, Wu Q, Zhu S, Li Y, Wu P, Zheng J, Shi H. RU.521 mitigates subarachnoid hemorrhage-induced brain injury via regulating microglial polarization and neuroinflammation mediated by the cGAS/STING/NF-κB pathway. Cell Commun Signal 2023; 21:264. [PMID: 37770901 PMCID: PMC10537158 DOI: 10.1186/s12964-023-01274-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/13/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND The poor prognosis of subarachnoid hemorrhage (SAH) is often attributed to neuroinflammation. The cGAS-STING axis, a cytoplasmic pathway responsible for detecting dsDNA, plays a significant role in mediating neuroinflammation in neurological diseases. However, the effects of inhibiting cGAS with the selective small molecule inhibitor RU.521 on brain injury and the underlying mechanisms after SAH are still unclear. METHODS The expression and microglial localization of cGAS following SAH were investigated with western blot analysis and immunofluorescent double-staining, respectively. RU.521 was administered after SAH. 2'3'-cGAMP, a second messenger converted by activated cGAS, was used to activate cGAS-STING. The assessments were carried out by adopting various techniques including neurological function scores, brain water content, blood-brain barrier permeability, western blot analysis, TUNEL staining, Nissl staining, immunofluorescence, morphological analysis, Morris water maze test, Golgi staining, CCK8, flow cytometry in the in vivo and in vitro settings. RESULTS Following SAH, there was an observed increase in the expression levels of cGAS in rat brain tissue, with peak levels observed at 24 h post-SAH. RU.521 resulted in a reduction of brain water content and blood-brain barrier permeability, leading to an improvement in neurological deficits after SAH. RU.521 had beneficial effects on neuronal apoptosis and microglia activation, as well as improvements in microglial morphology. Additionally, RU.521 prompted a shift in microglial phenotype from M1 to M2. We also noted a decrease in the production of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, and an increase in the level of the anti-inflammatory cytokine IL-10. Finally, RU.521 treatment was associated with improvements in cognitive function and an increase in the number of dendritic spines in the hippocampus. The therapeutic effects were mediated by the cGAS/STING/NF-κB pathway and were found to be abolished by 2'3'-cGAMP. In vitro, RU.521 significantly reduced apoptosis and neuroinflammation. CONCLUSION The study showed that SAH leads to neuroinflammation caused by microglial activation, which contributes to early brain injury. RU.521 improved neurological outcomes and reduced neuroinflammation by regulating microglial polarization through the cGAS/STING/NF-κB pathway in early brain injury after SAH. RU.521 may be a promising candidate for the treatment of neuroinflammatory pathology after SAH. Video Abstract.
Collapse
Affiliation(s)
- Jiang Shao
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yuxiao Meng
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Kaikun Yuan
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Qiaowei Wu
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Shiyi Zhu
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yuchen Li
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Pei Wu
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Jiaolin Zheng
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Xuefu Road 246#, Nangang District, Harbin, 150001, Heilongjiang Province, China.
| | - Huaizhang Shi
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|