1
|
Zhang X, Gao L, Wang J, Zhang W, Xu D, Wang Y, Liu T, Gao K, Ren Z, Ding Y. Study of the ameliorative effect of β-Bisabolene on ischemic stroke via COX-2 with the Keap1/Nrf2 and MAPK pathways. Eur J Pharmacol 2025; 1001:177773. [PMID: 40441590 DOI: 10.1016/j.ejphar.2025.177773] [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: 03/29/2025] [Revised: 05/08/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025]
Abstract
β-Bisabolene, a bioactive sesquiterpene extracted from myrrh-a substance utilized for ischemic stroke treatment-has known therapeutic potential. Nevertheless, the exact mechanisms underlying its beneficial effects on ischemic stroke, along with its potential targets, remain to be fully elucidated. This study aimed to investigate the therapeutic potential of β-Bisabolene in ameliorating ischemic stroke, identify its potential targets and interactions, and explore the underlying pathways involved. Bioinformatics analyses, including network pharmacology and analysis of GEO transcriptomic datasets, were performed to predict the potential targets and mechanisms of β-Bisabolene in ischemic stroke treatment. Preliminary computational validation was achieved through molecular similarity comparisons, molecular docking studies, and molecular dynamics simulations. In vitro experiments were then conducted to confirm the protective effects of β-Bisabolene on microglia injured by oxygen‒glucose deprivation and to validate the findings from the bioinformatics analyses. The results revealed that β-Bisabolene reduces the levels of the proinflammatory cytokines TNF-α, IL-1β and NO; decreases reactive oxygen species levels; and decreases the expression of COX-2, P38, ERK and Keap1. Moreover, it increases the expression of Arg-1, Nrf2, NQO1 and HO-1. These effects are associated with improved survival rates of oxygen-glucose deprivation-damaged microglia. In conclusion, β-Bisabolene may exert anti-inflammatory and antioxidant effects to ameliorate microglial injury induced by ischemic stroke by antagonizing COX-2 and mediating the MAPK signaling pathway and Keap1/Nrf2 pathway. This study provides valuable insights into the therapeutic potential of β-Bisabolene for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Xingfang Zhang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining, 810008, China
| | - Liang Gao
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining, 810008, China
| | - Jianv Wang
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining, 810008, China
| | - Wei Zhang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Dong Xu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yanhua Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Tianlong Liu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Kai Gao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhen Ren
- Department of Ultrasonography, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
Zhang X, Li H, Zhao Y, Zhao T, Wang Z, Tang Q. Neuronal Injury after Ischemic Stroke: Mechanisms of Crosstalk Involving Necroptosis. J Mol Neurosci 2025; 75:15. [PMID: 39903429 DOI: 10.1007/s12031-025-02313-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Ischemic stroke is a leading cause of disability and death worldwide, largely due to its increasing incidence associated with an aging population. This condition results from arterial obstruction, significantly affecting patients' quality of life and imposing a substantial economic burden on healthcare systems. While current treatments primarily focus on the rapid restoration of blood flow through thrombolytic therapy or surgical interventions, a limited understanding of neuronal injury mechanisms hampers the development of more effective treatments.This article explores the interplay among various cell death pathways-necroptosis, apoptosis, autophagy, ferroptosis, and pyroptosis-in the context of ischemic stroke to identify novel therapeutic targets. Each mode of cell death displays unique characteristics and roles post-stroke, and the activation of these pathways may vary across different animal models, complicating the translation of therapeutic strategies to clinical settings. Notably, the interaction between apoptosis and necroptosis is highlighted; inhibiting apoptosis might heighten the risk of necroptosis. Therefore, a balanced regulation of these pathways could promote enhanced neuronal survival.Additionally, we introduce PANoptosis, a form of cell death that encompasses pyroptosis, apoptosis, and necroptosis, emphasizing the complexity and potential therapeutic implications of these interactions. In summary, understanding the relationships among these cell death mechanisms in ischemic stroke is vital for developing new neuroprotective agents. Future research should aim for combinatorial interventions targeting multiple pathways to optimize treatment strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Xuanning Zhang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Hongyu Li
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Yaowei Zhao
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Tingting Zhao
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Zhihao Wang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Qiang Tang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China.
| |
Collapse
|
3
|
Zhang Y, Yang L, Gan Y, Zhao C, Zhou C, Chen J, Yin Y, Xia S, Yang H, Bao X, Zhang M, Xu Y, Li J. Benzydamine attenuates microglia-mediated neuroinflammation and ischemic brain injury by targeting cathepsin s. Int Immunopharmacol 2025; 146:113824. [PMID: 39700961 DOI: 10.1016/j.intimp.2024.113824] [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: 09/14/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Microglia, the primary immune cells of the central nervous system, play a crucial role in the neuroinflammatory processes following ischemic stroke. Targeting neuroinflammation is a promising strategy to enhance the outcomes of ischemic stroke. Benzydamine (BA), a well-known non-steroidal anti-inflammatory drug, has demonstrated potential in inhibiting pro-inflammatory cytokines across various disease models. However, the potential role of BA in microglial activation and post-stroke neuroinflammation remains unclear. Our study reveals that BA effectively suppresses the lipopolysaccharide (LPS)-stimulated pro-inflammatory responses of primary microglia, with high-dose BA (10 μM) suppressing LPS-induced inflammatory markers by up to 59.1 % in the mRNA levels of IL-1β. Furthermore, BA mitigated ischemic brain injury in experimental stroke mice. BA treatment also significantly attenuated neuroinflammatory responses and attenuates ischemic brain injury in experimental stroke mice. Further investigation revealed that BA reduces the release of the LPS-stimulated pro-inflammatory factors and activation of primary microglia by directly binding to and inhibiting the activity of cathepsin S (CTSS). In conclusion, our study identifies BA as a promising CTSS inhibitor with potential to suppress neuroinflammation following ischemic stroke. Our findings provide a theoretical basis for developing new neuroprotective strategies.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Lixuan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yonghui Gan
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Chenchen Zhao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Chao Zhou
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Jian Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yanping Yin
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Disease, Nanjing University, Nanjing 210008, China
| | - Haiyan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Disease, Nanjing University, Nanjing 210008, China
| | - Meijuan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Disease, Nanjing University, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China; Nanjing Neurology Clinical Medical Center, Nanjing, 210008, China.
| | - Jingwei Li
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
4
|
Li W, Feng X, Zhang M, Wang K, Huang K, Zhao Z, Xia M. CLEC7A Knockdown Alleviates Ischemic Stroke by Inhibiting Pyroptosis and Microglia Activation. J Integr Neurosci 2024; 23:219. [PMID: 39735961 DOI: 10.31083/j.jin2312219] [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: 04/15/2024] [Revised: 07/26/2024] [Accepted: 08/20/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Ischemic stroke (IS) is the leading cause of mortality worldwide. Herein, we aimed to identify novel biomarkers and explore the role of C-type lectin domain family 7 member A (CLEC7A) in IS. METHODS Differentially expressed genes (DEGs) were screened using the GSE106680, GSE97537, and GSE61616 datasets, and hub genes were identified through construction of protein-protein interaction networks. An IS model was established by middle cerebral artery occlusion and reperfusion (MCAO/R). Neural function was assessed using triphenyl tetrazolium chloride, hematoxylin-eosin, and terminal deoxynucleotidyl transferase-mediated nick-end labeling. A cell counting kit was used to detect cell viability following oxygen-glucose deprivation/reperfusion (OGD/R). Inflammatory factors were detected using enzyme-linked immunosorbent assay. The mRNA and protein expression levels were detected using reverse transcription-quantitative polymerase chain reaction and western blotting, respectively. RESULTS Fc fragment of Immunoglobulin G (IgG) receptor IIIa (FCGR3A), Fc fragment of Immunoglobulin E (IgE) receptor Ig (FCER1G), Complement component 5a receptor 1 (C5AR1), CLEC7A, Plasminogen activator, urokinase (PLAU), and C-C motif chemokine ligand 6 (CCL6) were identified as important hub genes, from which CLEC7A was selected as the primary subject of this study. The activation of microglia and pyroptosis were observed in MCAO/R model with increased levels of interleukin (IL)-1β, IL-18, tumor necrosis factor-α, and lactate dehydrogenase. CLEC7A knockdown was found to promote cell viability in BV2 cells and inhibiting pyroptosis in HT22 cells. CLEC7A knockdown in microglia also decreased infarct volume and neurological deficit scores, and alleviated injury and neuronal apoptosis in IS rats. CLEC7A knockdown inhibited pyroptosis and microglial activation in the MCAO/R model. A pyroptosis activator reversed the effect of CLEC7A knockdown on the viability of OGD/R-treated HT22 cells. CONCLUSION CLEC7A is a promising biomarker of IS. CLEC7A knockdown alleviates IS by inhibiting pyroptosis and microglial activation.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Xiaoli Feng
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Manyu Zhang
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Kangmeng Wang
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Kailai Huang
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Zhenqiang Zhao
- Department of Neurology, First Affiliated Hospital of Hainan Medical University, 570102 Haikou, Hainan, China
| | - Min Xia
- Department of Neurology, Hainan West Central Hospital, 571799 Danzhou, Hainan, China
| |
Collapse
|
5
|
Xu Y, Liu W, Ren L. Role of m6A RNA Methylation in Ischemic Stroke. Mol Neurobiol 2024; 61:6997-7008. [PMID: 38363537 DOI: 10.1007/s12035-024-04029-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Ischemic stroke is a prominent contributor to global morbidity and mortality rates. The intricate and diverse mechanisms underlying ischemia-reperfusion injury remain poorly comprehended. RNA methylation, an emerging epigenetic modification, plays a crucial role in regulating numerous biological processes, including immunity, DNA damage response, tumorigenesis, metastasis, stem cell renewal, adipocyte differentiation, circadian rhythms, cellular development and differentiation, and cell division. Among the various RNA modifications, N6-methyladenosine (m6A) modification stands as the most prevalent in mammalian mRNA. Recent studies have demonstrated the crucial involvement of m6A modification in the pathophysiological progression of ischemic stroke. This review aims to elucidate the advancements in ischemic stroke-specific investigations pertaining to m6A modification, consolidate the underlying mechanisms implicated in the participation of m6A modification during the onset of ischemic stroke, and deliberate on the potential of m6A modification as a viable therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Yayun Xu
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Wenqiang Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230000, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230000, China
| | - Lijie Ren
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| |
Collapse
|
6
|
Zheng H, Wu H, Wang D, Wang S, Ji D, Liu X, Gao G, Su X, Zhang Y, Ling Y. Research progress of prodrugs for the treatment of cerebral ischemia. Eur J Med Chem 2024; 272:116457. [PMID: 38704941 DOI: 10.1016/j.ejmech.2024.116457] [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: 03/21/2024] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
It is well-known that pharmacotherapy plays a pivotal role in the treatment and prevention of cerebral ischemia. Nevertheless, existing drugs, including numerous natural products, encounter various challenges when applied in cerebral ischemia treatment. These challenges comprise poor brain absorption due to low blood-brain barrier (BBB) permeability, limited water solubility, inadequate bioavailability, poor stability, and rapid metabolism. To address these issues, researchers have turned to prodrug strategies, aiming to mitigate or eliminate the adverse properties of parent drug molecules. In vivo metabolism or enzymatic reactions convert prodrugs into active parent drugs, thereby augmenting BBB permeability, improving bioavailability and stability, and reducing toxicity to normal tissues, ultimately aiming to enhance treatment efficacy and safety. This comprehensive review delves into multiple effective prodrug strategies, providing a detailed description of representative prodrugs developed over the past two decades. It underscores the potential of prodrug approaches to improve the therapeutic outcomes of currently available drugs for cerebral ischemia. The publication of this review serves to enrich current research progress on prodrug strategies for the treatment and prevention of cerebral ischemia. Furthermore, it seeks to offer valuable insights for pharmaceutical chemists in this field, offer guidance for the development of drugs for cerebral ischemia, and provide patients with safer and more effective drug treatment options.
Collapse
Affiliation(s)
- Hongwei Zheng
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Hongmei Wu
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Dezhi Wang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Sijia Wang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Dongliang Ji
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Xiao Liu
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Ge Gao
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Xing Su
- Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Yanan Zhang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Yong Ling
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China.
| |
Collapse
|