1
|
Yao H, Wu R, Du D, Ai F, Yang F, Li Y, Qi S. Flavonoids from Polypodium hastatum as neuroprotective agents attenuate cerebral ischemia/reperfusion injury in vitro and in vivo via activating Nrf2. Redox Rep 2025; 30:2440204. [PMID: 39702961 DOI: 10.1080/13510002.2024.2440204] [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] [Indexed: 12/21/2024] Open
Abstract
OBJECTIVES Cerebral ischemic stroke is a leading cause of death worldwide. Though timely reperfusion reduces the infarction size, it exacerbates neuronal apoptosis due to oxidative stress. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor regulating the expression of antioxidant enzymes. Activating Nrf2 gives a therapeutic approach to ischemic stroke. METHODS Herein we explored flavonoids identified from Polypodium hastatum as Nrf2 activators and their protective effects on PC12 cells injured by oxygen and glucose deprivation/restoration (OGD/R) as well as middle cerebral artery occlusion (MCAO) mice. RESULTS The results showed among these flavonoids, AAKR significantly improved the survival of PC12 cells induced by OGD/R and activated Nrf2 in a Keap1-dependent manner. Further investigations have disclosed AAKR attenuated oxidative stress, mitochondrial dysfunction and following apoptosis resulting from OGD/R. Meanwhile, activation of Nrf2 by AAKR was involved in the protective effects. Finally, it was found that AAKR could protect MCAO mice brains against ischemia/reperfusion injury via activating Nrf2. DISCUSSION This investigation could provide lead compounds for the discovery of novel Nrf2 activators targeting ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Huankai Yao
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Ruiqing Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Dan Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Fengwei Ai
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Feng Yang
- School of Stomatology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yan Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy & Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Suhua Qi
- School of Medical Technology & Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|
2
|
Ding YQ, Zhao D, Chen X, Yuan HM, Mao LJ. Effect of Huatuo Zaizao Pill on Neurological Function and Limb Motor Recovery in Ischemic Stroke Patients During Convalescence: An Open-Labelled, Randomized Controlled Trial. Chin J Integr Med 2025:10.1007/s11655-025-3928-4. [PMID: 40232598 DOI: 10.1007/s11655-025-3928-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2024] [Indexed: 04/16/2025]
Abstract
OBJECTIVE To evaluate the effects of Chinese patent medicine Huatuo Zaizao Pill (HTZZ) on neurological function and limb motor in ischemic stroke (IS) patients during convalescence. METHODS This is a prospective, open-labelled, randomized controlled trial. Patients with IS were recruited from the Neurology Department of Xiyuan Hospital of China Academy of Chinese Medical Sciences from May 2021 to June 2023. Eligible participants were randomly assigned to the HTZZ (40 cases) or control group (40 cases) at a ratio of 1:1. The HTZZ group was treated with oral HTZZ (8 g, thrice daily) combined with conventional treatment, while the control group received only conventional treatment. The treatment duration was 12 weeks. The primary outcome was the change in Modified Ashworth Scale (MAS) score from baseline to week 6 and 12. Secondary outcomes included changes in scores of National Institute of Health Stroke Scale (NIHSS), Fugl-Meyer Assessment (FM), and Barthel Index (BI) from baseline to week 6 and 12, as well as lipid indices after 12 weeks. All adverse events (AEs) were recorded and liver and kidney indices were evaluated. RESULTS A total of 72 patients completed the study (38 in the HTZZ group and 34 in the control group). Compared with the control group, the HTZZ group demonstrated significant improvements in MAS, NIHSS, FM, and BI scores following 6 and 12 weeks of treatment in both intent-to-treat and per-protocol analyses (all P<0.05). No significant differences were noted between groups in lipid indices, AEs, and liver and kidney dysfunction after 12 weeks (P>0.05). CONCLUSIONS HTZZ alleviated spasticity and enhanced neurological function and prognosis of IS patients during convalescence. However, further evaluation of HTZZ's effect on IS outcomes is warranted in clinical trials with larger sample sizes and extended observation periods. (Trial registration No. NCT04910256).
Collapse
Affiliation(s)
- Yan-Qiu Ding
- Graduate School of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dan Zhao
- Graduate School of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiao Chen
- Graduate School of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hui-Min Yuan
- Graduate School of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Li-Jun Mao
- Department of Neurology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
3
|
Zhou J, Li C, Yue Y, Kim YK, Park S. Multitarget Natural Compounds for Ischemic Stroke Treatment: Integration of Deep Learning Prediction and Experimental Validation. J Chem Inf Model 2025; 65:3309-3323. [PMID: 40084909 DOI: 10.1021/acs.jcim.5c00135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Ischemic stroke's complex pathophysiology demands therapeutic approaches targeting multiple pathways simultaneously, yet current treatments remain limited. We developed an innovative drug discovery pipeline combining a deep learning approach with experimental validation to identify natural compounds with comprehensive neuroprotective properties. Our computational framework integrated SELFormer, a transformer-based deep learning model, and multiple deep learning algorithms to predict NC bioactivity against seven crucial stroke-related targets (ACE, GLA, MMP9, NPFFR2, PDE4D, and eNOS). The pipeline encompassed IC50 predictions, clustering analysis, quantitative structure-activity relationship (QSAR) modeling, and uniform manifold approximation and projection (UMAP)-based bioactivity profiling followed by molecular docking studies and experimental validation. Analysis revealed six distinct NC clusters with unique molecular signatures. UMAP projection identified 11 medium-activity (6 < pIC50 ≤ 7) and 57 high-activity (pIC50 > 7) compounds, with molecular docking confirming strong correlations between binding energies and predicted pIC50 values. In vitro studies using NGF-differentiated PC12 cells under oxygen-glucose deprivation demonstrated significant neuroprotective effects of four high-activity compounds: feruloyl glucose, l-hydroxy-l-tryptophan, mulberrin, and ellagic acid. These compounds enhanced cell viability, reduced acetylcholinesterase activity and lipid peroxidation, suppressed TNF-α expression, and upregulated BDNF mRNA levels. Notably, mulberrin and ellagic acid showed superior efficacy in modulating oxidative stress, inflammation, and neurotrophic signaling. This study establishes a robust deep learning-driven framework for identifying multitarget natural therapeutics for ischemic stroke. The validated compounds, particularly mulberrin and ellagic acid, are promising for stroke treatment development. Our findings demonstrate the effectiveness of integrating computational prediction with experimental validation in accelerating drug discovery for complex neurological disorders.
Collapse
Affiliation(s)
- Junyu Zhou
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Department of Bioconvergence, Hoseo University, Asan 31499, South Korea
| | - Chen Li
- Department of Bioconvergence, Hoseo University, Asan 31499, South Korea
| | - Yu Yue
- Department of Bioconvergence, Hoseo University, Asan 31499, South Korea
| | - Yong Kwan Kim
- Department of Information and Communication Engineering, Hoseo University, Asan 31499, South Korea
| | - Sunmin Park
- Department of Bioconvergence, Hoseo University, Asan 31499, South Korea
- Dept. of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, Asan 31499, Korea
| |
Collapse
|
4
|
An Y, Huang L, Li J, Chen Z, Cai J, Wang B, Zhou Q. LncRNA-mRNA co-expression network in the mechanism of butylphthalide treatment for ischemic stroke. BMC Neurol 2025; 25:155. [PMID: 40211238 PMCID: PMC11984060 DOI: 10.1186/s12883-025-04032-8] [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: 05/24/2024] [Accepted: 01/10/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Butylphthalide has shown significant potential in the treatment of ischemic stroke, but its precise mechanisms of action remain unclear. Long non-coding RNAs (lncRNAs) and messenger RNAs (mRNAs) play crucial roles in the pathogenesis of ischemic stroke and may serve as potential therapeutic targets. This study investigated the effects of butylphthalide treatment on the lncRNA-mRNA co-expression network in ischemic stroke patients. METHODS Peripheral blood samples were collected from ischemic stroke patients treated with butylphthalide and from control subjects. mRNA and lncRNA expression profiles were obtained using microarray scanning, and differentially expressed lncRNAs (DElncRNAs) were validated by qRT-PCR. Target genes interacting with DElncRNAs were predicted using the miRTargetLink database. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed on both DElncRNAs and differentially expressed mRNAs (DEmRNAs). A protein-protein interaction (PPI) network was constructed for proteins encoded by DEmRNAs. Co-expression analysis, based on Pearson correlation coefficients, identified the top five mRNAs and lncRNAs with high connectivity. Finally, molecular docking was performed to investigate the binding interaction between butylphthalide and key mRNAs. RESULTS A total of 86 differentially expressed mRNAs (69 upregulated, 17 downregulated) and 35 DElncRNAs (all upregulated) were identified. DEmRNAs were primarily associated with pathways related to cell receptors, signal transduction, cell proliferation, migration, and glucose metabolism, while DElncRNAs were involved in processes such as embryonic development, neuronal connectivity, and energy metabolism. Co-expression analysis identified key mRNA nodes (SETD9, ZNF718, AOC2, MPND, ODF1) and lncRNA nodes (IDH2-DT, CLEC12A-AS1, CARD8-AS1, LINC01275, ZNF436-AS1). Molecular docking analysis suggested that MT-CO1, SETD9, and ZNF718 could be potential targets of butylphthalide. CONCLUSION Butylphthalide may exert its therapeutic effects by regulating the LncRNA-mRNA co-expression network, influencing energy metabolism and neuronal development. This provides new insights into its mechanism of action and potential therapeutic targets.
Collapse
Affiliation(s)
- Yangfang An
- Department of Neurology, Yiyang Central Hospital, Yiyang, Hunan, 413000, China
| | - Lingyun Huang
- Department of Neurology, Yiyang Central Hospital, Yiyang, Hunan, 413000, China
| | - Jun Li
- Department of Neurology, Yiyang Central Hospital, Yiyang, Hunan, 413000, China
| | - Zhuo Chen
- Department of Neurology, Yiyang Central Hospital, Yiyang, Hunan, 413000, China
| | - Jizhang Cai
- Department of Neurology, Yiyang Central Hospital, Yiyang, Hunan, 413000, China
| | - Biao Wang
- Department of Neurology, Yiyang Central Hospital, Yiyang, Hunan, 413000, China
| | - Qiong Zhou
- Department of Neurology, Yiyang Central Hospital, Yiyang, Hunan, 413000, China.
| |
Collapse
|
5
|
Gao J, Wei Z, Wang Z, Zhang Y. Global trends, disparities, and future projections of ischemic stroke burden attributed to low-fiber diets: An analysis based on GBD 2021. J Stroke Cerebrovasc Dis 2025; 34:108308. [PMID: 40209963 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/31/2025] [Accepted: 04/05/2025] [Indexed: 04/12/2025] Open
Abstract
OBJECTIVE This study aims to analyze the disease burden attributed to ischemic stroke related to low-fiber diets (IFD-IS) from 1990 to 2021, utilizing data from the Global Burden of Disease (GBD) database to identify trends and influencing factors of the disease burden. METHODS We conducted a comprehensive analysis of the number of deaths, Disability-Adjusted Life Years (DALYs), Years of Life Lost (YLLs), and Years Lost due to Disability (YLDs) associated with IFD-IS, covering various regions and populations. Statistical methods, including breakpoint regression, decomposition analysis, health inequality analysis, and forecasting analysis, were employed to assess trends and their correlation with the Sociodemographic Index (SDI). RESULTS The findings indicate that deaths, DALYs, YLLs, and YLDs related to IFD-IS have gradually increased globally, with a significantly higher burden in males compared to females. Notable regional disparities were observed, particularly with heavier burdens in China and sub-Saharan Africa. Higher SDI regions demonstrated better control over disease burdens, while aging and population growth were the primary factors contributing to the increase. Additionally, the trend of health inequality related to IFD-IS has slightly diminished, although it is projected that the disease burden will continue to rise by 2040. CONCLUSION This study underscores the necessity of implementing targeted public health interventions for IFD-IS, particularly in low SDI regions. Enhancing dietary education and resource allocation can significantly mitigate the rising trend of IFD-IS. Ongoing monitoring and research are crucial for formulating effective health policies to address this public health challenge.
Collapse
Affiliation(s)
- Jianhong Gao
- Hubei Minzu University, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Hubei, 445000, China; Health Medical Center, Hubei Minzu University, Enshi, Hubei, 445000, China
| | - Zhenhua Wei
- Hubei Minzu University, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Hubei, 445000, China; Health Medical Center, Hubei Minzu University, Enshi, Hubei, 445000, China
| | - Zhengyu Wang
- Health Medical Center, Hubei Minzu University, Enshi, Hubei, 445000, China
| | - Yun Zhang
- Department of Clinical Laboratory Center, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, Hubei Province, China.
| |
Collapse
|
6
|
Xu L, Wang P, Yang L, Liu Y, Li X, Yin Y, Lan C. Neurotrophic factor biomarkers for ischemic stroke diagnosis and mechanistic insights. Sci Rep 2025; 15:11906. [PMID: 40195336 PMCID: PMC11977241 DOI: 10.1038/s41598-025-86935-7] [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: 07/22/2024] [Accepted: 01/15/2025] [Indexed: 04/09/2025] Open
Abstract
Ischemic stroke (IS), a multifactorial disease resulting from the complex interplay of various environmental and genetic risk factors. Neurotrophic factors (NTFs) have a potential role in IS, but the exact mechanisms are unknown. The aim of this study was to identify biomarkers associated with the occurrence and development of NTFs and to analyze their potential mechanisms of action. In this study, we selected the intersection of neurotrophic factor genes, differentially expressed genes (DEGs) and key genes in the IS module based on IS-related datasets (GSE16561 and GSE58294). Machine learning screened out 5 biomarkers for IS diagnosis (MMP9, MARCKS, IGF2R, HECW2 and CYBRD1). GSEA results showed that different signaling pathways were activated in IS samples with high expression of different diagnostic genes. Furthermore, an immunological analysis was carried out, which demonstrated significant differences in the levels of activated B cells, neutrophils, and activated CD8 T cells between IS patients and normal samples. RT-qPCR results showed that there were significant differences in the expression of CYBRD1, MARCKS and MMP9 between IS and control patients. In conclusion, we identified 5 diagnostic markers that may be involved in the progression of IS, including MMP9, MARCKS, IGF2R, HECW2 and CYBRD1. Finally, differential expression of MMP9, MARCKS, and CYBRD1 was detected in peripheral blood samples from 15 IS and 5 normal cases. Our analysis could serve as a foundation for enhancing comprehension of the underlying molecular mechanisms governing the pathogenesis and progression of IS. The identified biomarkers might serve as targets for the development of novel diagnostic assays, enabling earlier detection of IS and potentially leading to more timely and effective treatment interventions.
Collapse
Affiliation(s)
- Liying Xu
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Pingzhi Wang
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| | - Lei Yang
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yinlian Liu
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xiangping Li
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yajie Yin
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Caiqin Lan
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| |
Collapse
|
7
|
González Torrecilla S, Delbrel A, Giacomino L, Meunier D, Sein J, Renaud L, Brige P, Garrigue P, Hak JF, Guillet B, Brunel H, Farjot G, Brochier T, Velly L. Long lasting argon neuroprotection in a non-human primate model of transient endovascular ischemic stroke. J Cereb Blood Flow Metab 2025; 45:643-654. [PMID: 39628320 PMCID: PMC11615904 DOI: 10.1177/0271678x241297798] [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: 04/04/2024] [Revised: 09/26/2024] [Accepted: 10/14/2024] [Indexed: 12/06/2024]
Abstract
In the past decade, noble gases have emerged as highly promising neuroprotective agents. Previous studies have demonstrated the efficacy of argon neuroprotection in rodent models of cerebral ischemia. The objective of the present pre-clinical study was to confirm the neuroprotective effect of argon in a non-human primate model of endovascular ischemic stroke. Thirteen adult Macaca mulatta were subjected to a focal cerebral ischemia induced by a transient (90 min) middle cerebral artery occlusion (tMCAO). The monkeys were randomly allocated to a control group (n = 8) and an argon group (n = 5). Pre-mixed gas (40-60 oxygen-argon) was applied 30 min after the onset of tMCAO to 30 min after reperfusion. Infarct volumes were measured from the MRI scans conducted 1 hour and 1 month after the reperfusion. A clinical neurological assessment was performed 24 hours and 1 month after tMCAO. Our results show that Argon dramatically reduced ischemic core volume after ischemia compared to the control group with a long-lasting improvement of post-stroke infarct volume at 1 month. In addition, the neurological scale suggests a better prognosis in argon-treated animals without reaching the significance threshold. These pre-clinical results in gyrencephalic non-human primates support the potential use of this therapeutic approach for future clinical studies.
Collapse
Affiliation(s)
- Sandra González Torrecilla
- Institut de Neurosciences de la Timone (INT), CNRS, Aix-Marseille Université, Marseille, France
- Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Alisée Delbrel
- Institut de Neurosciences de la Timone (INT), CNRS, Aix-Marseille Université, Marseille, France
- Department of Anesthesiology and Critical Care Medicine, Marseille University Hospital Timone, AP-HM, Marseille, France
| | - Laura Giacomino
- Department of Anesthesiology and Critical Care Medicine, Marseille University Hospital Timone, AP-HM, Marseille, France
| | - David Meunier
- Institut de Neurosciences de la Timone (INT), CNRS, Aix-Marseille Université, Marseille, France
| | - Julien Sein
- Institut de Neurosciences de la Timone (INT), CNRS, Aix-Marseille Université, Marseille, France
- Centre Européen de Recherche en Imagerie Médicale (CERIMED), Aix-Marseille University, CNRS, Marseille, France
| | - Luc Renaud
- Institut de Neurosciences de la Timone (INT), CNRS, Aix-Marseille Université, Marseille, France
| | - Pauline Brige
- Institut de Neurosciences de la Timone (INT), CNRS, Aix-Marseille Université, Marseille, France
- Centre Européen de Recherche en Imagerie Médicale (CERIMED), Aix-Marseille University, CNRS, Marseille, France
| | - Philippe Garrigue
- Centre Européen de Recherche en Imagerie Médicale (CERIMED), Aix-Marseille University, CNRS, Marseille, France
- Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille University, INSERM, INRAE, Marseille, France
| | - Jean Francois Hak
- Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille University, INSERM, INRAE, Marseille, France
- Department of Neuroradiology, Marseille University Hospital Timone, AP-HM, Marseille, France
| | - Benjamin Guillet
- Centre Européen de Recherche en Imagerie Médicale (CERIMED), Aix-Marseille University, CNRS, Marseille, France
- Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille University, INSERM, INRAE, Marseille, France
| | - Hervé Brunel
- Department of Neuroradiology, Marseille University Hospital Timone, AP-HM, Marseille, France
| | | | - Thomas Brochier
- Institut de Neurosciences de la Timone (INT), CNRS, Aix-Marseille Université, Marseille, France
| | - Lionel Velly
- Institut de Neurosciences de la Timone (INT), CNRS, Aix-Marseille Université, Marseille, France
- Department of Anesthesiology and Critical Care Medicine, Marseille University Hospital Timone, AP-HM, Marseille, France
| |
Collapse
|
8
|
Zeng Q, Li H, Yang Z, Zhang Z, Zhang M, Yang F, Gao P, Huangfu X, Fang Y. Potential edaravone/benzocyclopentenone derivatives alleviate cerebral ischemia reperfusion injury as neuroprotective agents. Bioorg Chem 2025; 157:108288. [PMID: 39986107 DOI: 10.1016/j.bioorg.2025.108288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Based on the scaffold of edaravone, a clinically approved neuroprotective agent, a series of edaravone/benzocyclopentenone hybrid derivatives were designed, synthesized and evaluated for their biological activities in vitro and in vivo. Most of the compounds demonstrated promising neuroprotective effects, with derivatives containing benzofuranone or indanone as core moiety showing particularly strong activity. Among all derivatives, 17 compounds exhibited significantly improved neuronal cell viabilities compared to edaravone in an OGD/R model with rat primary neuronal cells, along with favorable safety profiles and blood-brain barrier permeability. Notably, compound 13, which includes a fluoro-substituted benzofuranone fragment, displayed the most potent neuroprotective effect in vitro and effectively reduced cerebral infarct area in vivo.
Collapse
Affiliation(s)
- Qing Zeng
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Huilan Li
- National Engineering Research Center for Manufacturing Technology of TCM Solid Preparation, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Zunhua Yang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ziwei Zhang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Mai Zhang
- National Engineering Research Center for Manufacturing Technology of TCM Solid Preparation, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Fukang Yang
- National Engineering Research Center for Manufacturing Technology of TCM Solid Preparation, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Puyuan Gao
- National Engineering Research Center for Manufacturing Technology of TCM Solid Preparation, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Xian Huangfu
- National Engineering Research Center for Manufacturing Technology of TCM Solid Preparation, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Yuanying Fang
- National Engineering Research Center for Manufacturing Technology of TCM Solid Preparation, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| |
Collapse
|
9
|
Nagase T, Yasuhara T, Kin K, Sasada S, Kawauchi S, Yabuno S, Sugahara C, Hirata Y, Miyake H, Sasaki T, Kawai K, Tanimoto S, Saijo T, Tanaka S. Therapeutic effects of intracerebral transplantation of human modified bone marrow-derived stromal cells (SB623) with voluntary and forced exercise in a rat model of ischemic stroke. Exp Neurol 2025; 386:115145. [PMID: 39805465 DOI: 10.1016/j.expneurol.2025.115145] [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: 09/29/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Ischemic stroke results in significant long-term disability and mortality worldwide. Although existing therapies, such as recombinant tissue plasminogen activator and mechanical thrombectomy, have shown promise, their application is limited by stringent conditions. Mesenchymal stem cell (MSC) transplantation, especially using SB623 cells (modified human bone marrow-derived MSCs), has emerged as a promising alternative, promoting neurogenesis and recovery. This study evaluated the effects of voluntary and forced exercise, alone and in combination with SB623 cell transplantation, on neurological and psychological outcomes in a rat model of ischemic stroke. Male Wistar rats that had undergone middle cerebral artery occlusion (MCAO) were divided into six groups: control, voluntary exercise (V-Ex), forced exercise (F-Ex), SB623 transplantation, SB623 + V-Ex, and SB623 + F-Ex. Voluntary exercise was facilitated using running wheels, while forced exercise was conducted on treadmills. Neurological recovery was assessed using the modified neurological severity score (mNSS). Psychological symptoms were evaluated through the open field test (OFT) and forced swim test (FST), and neurogenesis was assessed via BrdU labeling. Both exercise groups exhibited significant changes in body weight post-MCAO. Both exercises enhanced the treatment effect of SB623 transplantation. The forced exercise showed a stronger treatment effect on ischemic stroke than voluntary exercise alone, and the sole voluntary exercise improved depression-like behavior. The SB623 + F-Ex group demonstrated the greatest improvements in motor function, infarct area reduction, and neurogenesis. The SB623 + V-Ex group was most effective in alleviating depression-like behavior. Future research should optimize these exercise protocols and elucidate the underlying mechanisms to develop tailored rehabilitation strategies for stroke patients.
Collapse
Affiliation(s)
- Takayuki Nagase
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kyohei Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Susumu Sasada
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Satoru Yabuno
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Chiaki Sugahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yuichi Hirata
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Hayato Miyake
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Koji Kawai
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Shun Tanimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Tomoya Saijo
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Shota Tanaka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| |
Collapse
|
10
|
Liu X, Wang W, Nie Q, Liu X, Sun L, Ma Q, Zhang J, Wei Y. The Role and Mechanisms of Ubiquitin-Proteasome System-Mediated Ferroptosis in Neurological Disorders. Neurosci Bull 2025; 41:691-706. [PMID: 39775589 PMCID: PMC11979074 DOI: 10.1007/s12264-024-01343-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/29/2024] [Indexed: 01/11/2025] Open
Abstract
Ferroptosis is a form of cell death elicited by an imbalance in intracellular iron concentrations, leading to enhanced lipid peroxidation. In neurological disorders, both oxidative stress and mitochondrial damage can contribute to ferroptosis, resulting in nerve cell dysfunction and death. The ubiquitin-proteasome system (UPS) refers to a cellular pathway in which specific proteins are tagged with ubiquitin for recognition and degradation by the proteasome. In neurological conditions, the UPS plays a significant role in regulating ferroptosis. In this review, we outline how the UPS regulates iron metabolism, ferroptosis, and their interplay in neurological diseases. In addition, we discuss the future application of small-molecule inhibitors and identify potential drug targets. Further investigation into the mechanisms of UPS-mediated ferroptosis will provide novel insights and strategies for therapeutic interventions and clinical applications in neurological diseases.
Collapse
Affiliation(s)
- Xin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Wei Wang
- Cancer Biology Institute, Baotou Medical College, Baotou, 014010, China
| | - Qiucheng Nie
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xinjing Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Lili Sun
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qiang Ma
- Cancer Biology Institute, Baotou Medical College, Baotou, 014010, China
| | - Jie Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Yiju Wei
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
11
|
Song L, Wu Y, Yin L, Duan Y, Hua J, Rong M, Liu K, Yin J, Ma D, Zhang C, Xiao B, Ma C. Hydroxysafflower yellow A alleviates the inflammatory response in astrocytes following cerebral ischemia by inhibiting the LCN2/STAT3 feedback loop. Metab Brain Dis 2025; 40:161. [PMID: 40172584 PMCID: PMC11965183 DOI: 10.1007/s11011-025-01581-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Lipocalin-2 (LCN2), an acute phase protein mainly expressed in astrocytes (Ast), is closely related to the production of inflammatory cytokines following ischemic stroke. During the pathophysiological process of ischemic stroke, the Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway is activated. Despite evidence suggesting some link between the two, the relationship between the JAK2/STAT3 signaling pathway and the LCN2 expression in Ast following brain ischemia is incompletely understood. Hydroxysafflower yellow A (HSYA), an active ingredient found in Carthamus tinctorius L flowers, has been demonstrated to effectively mitigate cerebral ischemia via its anti-inflammatory effect. However, whether HSYA mitigates the neuroinflammatory damage after ischemic stroke by disrupting the interaction between the JAK2/STAT3 signaling pathway and LCN2 in Ast is unknown. Focusing on these two scientific questions, we established an in vivo middle cerebral artery occlusion/reperfusion (MCAO/R) rat model and in vitro primary astrocyte oxygen glucose deprivation/reperfusion (OGD/R) model. In vivo results showed that HSYA treatment alleviated nerve damage and inhibited the expression of LCN2 and inflammatory factors in Ast. In vitro results showed after OGD/R the expression of LCN2 and inflammatory cytokines increased and the JAK2/STAT3 was activated in Ast. Meanwhile, after OGD/R the JAK2/STAT3 activation in Ast increased LCN2 expression, and the inhibition of LCN2 expression by HSYA decreased the JAK2/STAT3 activation in Ast. These findings suggest that there is an interaction between the LCN2 and JAK2/STAT3 in Ast after ischemic stroke, which can enhance the inflammatory factors and exacerbate neuroinflammatory injury. Therefore, we conclude that HSYA may inhibit the LCN2/STAT3 loop in Ast, thereby mitigating neuroinflammation after cerebral ischemia.
Collapse
Affiliation(s)
- Lijuan Song
- Department of Physiology, Shanxi Medical University, Taiyuan, China
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Neurosurgery, Sinopharm Tongmei General Hospital, Datong, China
| | - Yige Wu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Lijun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Yanzhe Duan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Jianlin Hua
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Mengwei Rong
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Kexin Liu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Junjun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Dong Ma
- Department of Neurosurgery, Sinopharm Tongmei General Hospital, Datong, China
| | - Ce Zhang
- Department of Physiology, Shanxi Medical University, Taiyuan, China.
| | - Baoguo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| | - Cungen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China.
- Department of Neurosurgery, Sinopharm Tongmei General Hospital, Datong, China.
| |
Collapse
|
12
|
Li D, Huo X, Shen L, Qian M, Wang J, Mao S, Chen W, Li R, Zhu T, Zhang B, Liu K, Wu F, Bai Y. Astrocyte heterogeneity in ischemic stroke: Molecular mechanisms and therapeutic targets. Neurobiol Dis 2025; 209:106885. [PMID: 40139279 DOI: 10.1016/j.nbd.2025.106885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Ischemic stroke is one of the major causes of death and disability in adults, bringing a significant economic burden to the society and families. Despite significant advancements in stroke treatment, focusing solely on neurons is insufficient for improving disease progression and prognosis. Astrocytes are the most ubiquitous cells in the brain, and they undergo morphological and functional changes after brain insults, which has been known as astrocyte reactivity. Transcriptomics have shown that reactive astrocytes (RA) are heterogeneous, and they can be roughly classified into neurotoxic and neuroprotective types, thereby affecting the development of central nervous system (CNS) diseases. However, the relationship between stroke and reactive astrocyte heterogeneity has not been fully elucidated, and regulating the heterogeneity of astrocytes to play a neuroprotective role may provide a new perspective for the treatment of stroke. Here we systematically review current advancements in astrocyte heterogeneity following ischemic stroke, elucidate the molecular mechanisms underlying their activation, and further summarize promising therapeutic agents and molecular targets capable of modulating astrocyte heterogeneity.
Collapse
Affiliation(s)
- Daxing Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xinchen Huo
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ling Shen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Minjie Qian
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jindou Wang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shijie Mao
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wenjing Chen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Runheng Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Tianhao Zhu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Beicheng Zhang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Kunxuan Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Feifei Wu
- Laboratory for Human Anatomy, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Ying Bai
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
13
|
Qin R, Xu W, Qin Q, Liang X, Lai X, Xie M, Chen L. Identification of NETs-related genes as diagnostic biomarkers in ischemic stroke using RNA sequencing and single-cell analysis. Mamm Genome 2025:10.1007/s00335-025-10117-z. [PMID: 40107980 DOI: 10.1007/s00335-025-10117-z] [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: 12/03/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025]
Abstract
Neutrophil extracellular traps (NETs) are increasingly recognized for their involvement in ischemic stroke (IS), yet their precise contribution to IS outcomes is not fully understood. This study aims to elucidate the role of NETs in IS progression and identify potential biomarkers and therapeutic targets. In this study, mice were subjected to middle cerebral artery occlusion (MCAO). RNA sequencing was conducted on brain tissue samples to identify differentially expressed genes (DEGs) using the "limma" package. The diagnostic potential of these biomarkers was assessed using receiver operating characteristic (ROC) curve analysis. Additionally, single-cell RNA sequencing data were analyzed with the Seurat package to further investigate the cellular dynamics. We identified DEGs, and NETs-related genes associated with IS progression. Specifically, Ceacam3, Tnf, Selp, and Fcgr4 were found to be upregulated in MCAO samples, exhibiting diagnostic value as biomarkers for IS. Immune infiltration analysis indicated associations between these genes and various immune cell types. Gene Set Enrichment Analysis (GSEA) revealed their involvement in IS-related pathways, including ferroptosis, IL-17 signaling, leukocyte transendothelial migration, necroptosis, and NETs formation. Single-cell data confirmed the expression of Tnf, Selp, and Fcgr4 in neutrophils. CellChat analysis uncovered key cell-cell interactions in IS, emphasizing the role of neutrophils in communicating with microglia and T cells via the JAM pathway, with Thbs1 and Cd47 as key mediators. The findings provide insights into the cellular and molecular mechanisms underlying IS and may pave the way for novel therapeutic strategies targeting NETs in IS patients.
Collapse
Affiliation(s)
- Rongxing Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Wei Xu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- National Center for International Research of Biological Targeting Diagnosis and Therapy (Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research), Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Qingchun Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- National Center for International Research of Biological Targeting Diagnosis and Therapy (Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research), Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Xiaojun Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xinyu Lai
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Minshan Xie
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Li Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
- National Center for International Research of Biological Targeting Diagnosis and Therapy (Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research), Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| |
Collapse
|
14
|
Lin M, Wu H, Wan X, Liu N, Jiang Y, Sheng Y, Wang J, Xu H, Xue J, Qin Z, Wang Y. Mito-Apocynin Protects Against Kainic Acid-Induced Excitotoxicity by Ameliorating Mitochondrial Impairment. Mol Neurobiol 2025:10.1007/s12035-025-04827-3. [PMID: 40095344 DOI: 10.1007/s12035-025-04827-3] [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: 06/06/2024] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Neurodegenerative diseases represent significant global health challenges, with rising incidence rates. A substantial body of evidence indicates that excitotoxicity may be a critical target in the context of these diseases. However, effective pharmacological interventions aimed at mitigating excitotoxicity remain elusive. This study aimed to elucidate the neuroprotective effects and mechanisms of the mitochondrion-targeted NOX inhibitor, mito-apocynin, in the context of kainic acid (KA)-induced excitotoxicity. Our findings demonstrate that KA disrupts mitochondrial morphology, leading to impaired energy metabolism and mitochondrial dysfunction. Western blotting experiments revealed that KA compromises mitochondrial quality control. Additionally, Nissl staining and CCK8 assays indicated that mito-apocynin (administered at 75 μg/kg in vivo and 1 μM in vitro) significantly reduced neuronal death resulting from KA-induced excitotoxic damage in both in vivo and in vitro models. Furthermore, mito-apocynin improved neurobehavioral deficits induced by KA and mitigated mitochondrial dysfunction observed in vitro. Notably, mito-apocynin significantly reversed the KA-induced increase in NOX4 levels within the striatal mitochondria, reduced the ratio of phosphorylated DRP1 (Ser616) to total DRP1, and enhanced the expression of PGC-1α, PINK1, and Parkin proteins throughout the total striatum. In summary, mito-apocynin alleviates oxidative stress, preserves normal mitochondrial function and energy metabolism, and promotes mitochondrial quality control by modulating NOX expression in mitochondria, thereby reducing KA-induced excitotoxic damage.
Collapse
Affiliation(s)
- Miaomiao Lin
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Huanchen Wu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Xiaorui Wan
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Na Liu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Yiyue Jiang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Yichao Sheng
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Jing Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Haidong Xu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Jie Xue
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Zhenghong Qin
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Yan Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
15
|
Song W, Teng L, Wang H, Pang R, Liang R, Zhu L. Exercise preconditioning increases circulating exosome miR-124 expression and alleviates apoptosis in rats with cerebral ischemia-reperfusion injury. Brain Res 2025; 1851:149457. [PMID: 39824375 DOI: 10.1016/j.brainres.2025.149457] [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: 10/05/2024] [Revised: 12/22/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
OBJECTIVES Exercise as a non-pharmacological intervention can exert beneficial effects directly through exosomes crossing the blood-brain barrier and reduce apoptosis after cerebral ischaemia/reperfusion injury (CI/RI). miRNA-124 (miR-124) is present in exosomes and plays an important role in regulating cerebral neurological activity; however, the mechanism of the relationship between exercise and the activity of exosomes and apoptosis after CI/RI remains unclear. Therefore, the present study investigated the effects of exercise preconditioning on CI/RI from the perspective of exosomal miR-124 and apoptosis. METHODS The middle cerebral artery occlusion/reperfusion (MCAO/R) model was established by blocking the middle cerebral artery, and a motorized running wheel was chosen as the method of exercise preconditioning for rats, the morphology, particle concentration and particle size distribution of the exosome samples were identified at the 6 h, 12 h, and 24 h time points. RT-PCR, western blotting, immunohistochemistry, TUNEL staining, TTC staining and mNSS scores were used to investigate the effects of exercise preconditioning on apoptosis in MCAO/R rats. RESULTS The results showed exercise reduced neurological dysfunction and infarct size, increased the content of plasma exocrine miR-124 at 24 h, which inhibited the expression of STAT3, increased the expression of the anti-apoptotic BCL-2, and decreased the expression of the pro-apoptotic BAX, thereby reducing apoptosis. CONCLUSIONS Our findings indicated that exercise preconditioning can enhance the anti-apoptotic capacity of tissues in the rat ischemic penumbra and reduce apoptosis after CI/RI via the exosomal miR-124, STAT3, BCL-2/BAX pathway.
Collapse
Affiliation(s)
- Wenjing Song
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Lili Teng
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Haoran Wang
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Ruifeng Pang
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Runyu Liang
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Luwen Zhu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150000, China.
| |
Collapse
|
16
|
Qian Q, Lyu H, Wang W, Wang Q, Li D, Liu X, He Y, Shen M. Combined transcriptomic and proteomic analyses reveal relevant myelin features in mice with ischemic stroke. Funct Integr Genomics 2025; 25:64. [PMID: 40085348 PMCID: PMC11909235 DOI: 10.1007/s10142-025-01573-6] [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: 06/01/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Ischemic stroke (IS), a leading cause of global disability and mortality, is characterized by white matter damage and demyelination. Despite advances, the molecular mechanisms driving post-IS myelin pathology remain poorly understood, limiting therapeutic development. This study investigates key myelin-related genes (MRGs) and their regulatory networks to identify novel therapeutic targets. A transient middle cerebral artery occlusion (MCAO) model was established in C57BL/6 mice, with brain tissues collected at four timepoints (Sham0D, MCAO0D, MCAO7D, MCAO14D). Transcriptomic and proteomic sequencing were performed, followed by soft clustering (Mfuzz), functional enrichment (GO/KEGG), and ROC analysis to identify key MRGs. Competing endogenous RNA (ceRNA) networks were constructed, and drug prediction was conducted using the Comparative Toxicogenomics Database (CTD) and molecular docking. Expression validation was performed via qRT-PCR and Western blot. Integrated multi-omics analysis identified Wasf3 and Slc25a5 as key MRGs, enriched in mitochondrial respiration, calcium metabolism, and cytoskeletal regulation. The AUC values of the one-to-one model scores were all greater than 0.7, suggesting that Wasf3 and Slc25a5 were able to effectively discriminate between samples from different time points. A ceRNA network revealed critical interactions, including the Wasf3-mmu-miR-423-5p-H19 axis, linking apoptosis and myelin dysfunction. Drug prediction highlighted valproic acid (VPA) as a high-affinity binder for both genes (binding energies: - 4.2 and - 4.7 kcal/mol), suggesting its potential as a therapeutic candidate for IS. Experimental validation confirmed significant downregulation of Wasf3 mRNA (p < 0.01) and protein (p = 0.069) post-IS, while Slc25a5 showed no significant changes, potentially due to sample size limitations. This study establishes Wasf3 and Slc25a5 as pivotal regulators of post-IS myelin pathology and proposes VPA as a promising therapeutic candidate to enhance remyelination. The findings underscore the utility of multi-omics approaches in bridging molecular mechanisms to clinical translation, offering new strategies for IS diagnosis and treatment.
Collapse
Affiliation(s)
- Qiuyang Qian
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China
| | - Hao Lyu
- Department of Neurosurgery, Shenzhen Second People'S Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wei Wang
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China
| | - Qiwen Wang
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China
| | - Desheng Li
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China
| | - Xiaojia Liu
- Department of Neurosurgery, Shenzhen Second People'S Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, China
| | - Yi He
- Department of Neurosurgery, Shenzhen Second People'S Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, China
| | - Mei Shen
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China.
| |
Collapse
|
17
|
Du J, Liang X, Wang D, Wang Z, Shen R. Mechanism of KDM4A in Regulating Microglial Polarization in Ischemic Stroke. Appl Biochem Biotechnol 2025:10.1007/s12010-025-05207-2. [PMID: 40080374 DOI: 10.1007/s12010-025-05207-2] [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] [Accepted: 12/24/2024] [Indexed: 03/15/2025]
Abstract
Microglia polarization plays important roles in inflammatory processes after ischemic stroke. This study aimed to explore the mechanism of lysine-specific histone demethylase 4 (KDM4A) in microglia polarization after ischemic stroke. The mouse model was established using middle cerebral artery occlusion/reperfusion (MCAO/R) and the cell model was established by oxygen-glucose deprivation/reperfusion (OGD/R). The neurological deficits and brain tissue injury were evaluated. The biomarkers of microglia were determined. Levels of KDM4A/mouse double minute-2 homolog (MDM2)/C1q/TNF-related protein-3 (CTRP3) were measured. Inflammatory cytokines were quantified. The impact of KDM4A on microglia polarization was assessed. The enrichment of KDM4A or histone 3 lysine 9 trimethylation (H3K9me3) on the MDM2 promoter was analyzed. The ubiquitination and protein levels of CTRP3 after MG132 and cycloheximide treatment were determined. Results showed that KDM4A and MDM2 were upregulated while CTRP3 was downregulated. KDM4A downregulation alleviated neurological dysfunction, rescued motor capacity, reduced inflammatory infiltration, suppressed microglia activation, and promoted M2 polarization. KDM4A inhibited the enrichment of H3K9me3 on the MDM2 promoter, increasing MDM2 expression and downregulating CTRP3 expression via ubiquitination and degradation. MDM2 overexpression or CTRP3 downregulation averted the promotive role of silencing KDM4A in microglia polarization. In conclusion, KDM4A promotes microglia polarization to aggravate ischemic stroke via the MDM2/CTRP3 axis.
Collapse
Affiliation(s)
- Jingliang Du
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China
| | - Xianyang Liang
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China
| | - Denghui Wang
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China
| | - Zhen Wang
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China
| | - Ruile Shen
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24, Jinghua Road, Jianxi District, Luoyang, 471003, Henan Province, China.
| |
Collapse
|
18
|
Zheng Z, Wang R, Zhao Y, Zhang P, Xie D, Peng S, Li R, Zhang J. Salidroside Derivative SHPL-49 Exerts Anti-Neuroinflammatory Effects by Modulating Excessive Autophagy in Microglia. Cells 2025; 14:425. [PMID: 40136674 PMCID: PMC11941147 DOI: 10.3390/cells14060425] [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: 01/29/2025] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
The neuroinflammation triggered by cellular demise plays a pivotal role in ameliorating the injury associated with ischemic stroke, which represents a significant global burden of mortality and disability. The compound SHPL-49, a derivative of rhodioloside, was discovered by our research team and has previously demonstrated neuroprotective effects in rats with ischemic stroke. This study aimed to elucidate the underlying mechanisms of SHPL-49's protective effects. Preliminary investigations revealed that SHPL-49 effectively alleviates PMCAO-induced neuroinflammation. Further studies indicated that SHPL-49 downregulates the expression of the lysosomal protein LAMP-2 and reduces lysosomal activity, impeding the fusion of lysosomes and autophagosomes, thus inhibiting excessive autophagy and increasing the expression levels of the autophagy proteins LC3-II and P62. Furthermore, SHPL-49 effectively reverses the NF-κB nuclear translocation induced by the autophagy inducer rapamycin, significantly lowering the expression levels of the inflammatory factors IL-6, IL-1β, and iNOS. In a co-culture system of BV2 and PC12 cells, SHPL-49 enhanced PC12 cell viability by inhibiting excessive autophagy in BV2 cells and reducing the ratio of apoptotic proteins Bax and BCL-2. The overall findings suggest that SHPL-49 exerts its neuroprotective effects through the inhibition of excessive autophagy and the suppression of the NF-κB signaling pathway in microglia, thereby attenuating neuroinflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jiange Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional, Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| |
Collapse
|
19
|
Liu K, Ji Y, Xie Y, Wang C, Zhou J, Wei Z, Wang X, Zheng X, Cen Y, Zhang F, Xu B. Discovery of Isobenzofuran-1(3 H)-one Derivatives as Selective TREK-1 Inhibitors with In Vitro and In Vivo Neuroprotective Effects. J Med Chem 2025; 68:5804-5823. [PMID: 40040241 DOI: 10.1021/acs.jmedchem.4c03146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
TREK-1 regulates neuronal excitability and neuronal cell apoptosis, and inhibition of TREK-1 is a potential strategy to prevent cell death and achieve neuroprotection in an ischemic stroke. In this work, a series of novel isobenzofuran-1(3H)-one derivatives were designed and synthesized as TREK-1 inhibitors, and extensive structure-activity relationships led to the discovery of potent and selective TREK-1 inhibitors having IC50 values of a low micromolar level. Among them, Cpd8l potently and selectively inhibited TREK-1 (IC50 = 0.81 μM, selectivity >30 fold over other K+, Na+, and TRP channels). Cpd8l remarkably reduced the neuron death in the OGD/R-induced cortical neuronal injury model, while adenovirus silencing TREK-1 reduced its neuroprotective effect. Furthermore, Cpd8l could effectively ameliorate brain injury in MCAO/R model mice. Collectively, this work demonstrates that Cpd8l may serve as a novel lead compound to develop a highly potent and selective TREK-1 inhibitor for ischemic stroke treatment.
Collapse
Affiliation(s)
- Kaiyue Liu
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yunyun Ji
- The Affiliated Nanjing Pukou Traditional Chinese Medicine Hospital, Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu, China
| | - Yiming Xie
- The Affiliated Nanjing Pukou Traditional Chinese Medicine Hospital, Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu, China
| | - Chengyan Wang
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jie Zhou
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ziyi Wei
- The Affiliated Nanjing Pukou Traditional Chinese Medicine Hospital, Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu, China
| | - Xiaoyu Wang
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaotong Zheng
- The Affiliated Nanjing Pukou Traditional Chinese Medicine Hospital, Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu, China
| | - Yao Cen
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Fan Zhang
- The Affiliated Nanjing Pukou Traditional Chinese Medicine Hospital, Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu, China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Normal University, Guilin 541004, China
| | - Bailing Xu
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
20
|
Wu J, Li Z, Zhao J, Zhang T, Zhang J, Lv Q, Li J, Wang G, Han J, Zou Z. Atypical sulfur-containing physalin from Physalis minima and protective effect against ischemia-reperfusion injury. PHYTOCHEMISTRY 2025; 235:114478. [PMID: 40086499 DOI: 10.1016/j.phytochem.2025.114478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Four previously undescribed physalins (1-4), along with six known ones (5-10) were isolated and identified from the whole plants of Physalis minima L., a medicinal and edible plant traditionally used in southwest China. Their structures were established through comprehensive spectroscopic analyses, including high-resolution electrospray ionization mass spectrometry and 1D/2D nuclear magnetic resonance spectroscopy. Moreover, the absolute configurations of 1-3, 5 and 7 were examined by X-ray diffraction analyses. Compound 1, an undescribed sulfur-containing physalin, exhibited the most protective effect against oxygen-glucose deprivation/reperfusion (OGD/R)-stimulated ischemia-reperfusion (I/R) injury in PC12 cells. Meanwhile, compound 1 was found to reduce the inflammatory response, with mechanistic studies indicating that it decreased pyroptosis-associated proteins, such as cleaved-caspase1, NLRP3, and GSDMD N-terminus. Importantly, GSDMD knockdown significantly reversed the protective effects of compound 1, highlighting the involvement of pyroptosis in the compound's protective mechanism against OGD/R-induced I/R injury in PC12 cells in vitro.
Collapse
Affiliation(s)
- Jiangping Wu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wannan Medical College, Wuhu 241002, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Zixu Li
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wannan Medical College, Wuhu 241002, China
| | - Jianping Zhao
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, MS 38677, USA
| | - Tao Zhang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Jun Zhang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wannan Medical College, Wuhu 241002, China
| | - Qiuyue Lv
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wannan Medical College, Wuhu 241002, China
| | - Jiangfei Li
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wannan Medical College, Wuhu 241002, China
| | - Guodong Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wannan Medical College, Wuhu 241002, China
| | - Jun Han
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wannan Medical College, Wuhu 241002, China; Wuhu Modern Technology Research and Development Center of Chinese herbal Medicines and Functional Foods, Anhui College of Traditional Chinese Medicine, Wuhu 241002, China.
| | - Zhongmei Zou
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
21
|
Srikanth Y, Reddy DH, Anusha VL, Dumala N, Viswanadh MK, Chakravarthi G, Nalluri BN, Yadagiri G, Ramakrishna K. Unveiling the Multifaceted Pharmacological Actions of Indole-3-Carbinol and Diindolylmethane: A Comprehensive Review. PLANTS (BASEL, SWITZERLAND) 2025; 14:827. [PMID: 40094833 PMCID: PMC11902694 DOI: 10.3390/plants14050827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Cruciferae family vegetables are remarkably high in phytochemicals such as Indole-3-carbinol (I3C) and Diindolylmethane (DIM), which are widely known as nutritional supplements. I3C and DIM have been studied extensively in different types of cancers like breast, prostate, endometrial, colorectal, gallbladder, hepatic, and cervical, as well as cancers in other tissues. In this review, we summarized the protective effects of I3C and DIM against cardiovascular, neurological, reproductive, metabolic, bone, respiratory, liver, and immune diseases, infections, and drug- and radiation-induced toxicities. Experimental evidence suggests that I3C and DIM offer protection due to their antioxidant, anti-inflammatory, antiapoptotic, immunomodulatory, and xenobiotic properties. Apart from the beneficial effects, the present review also discusses the possible toxicities of I3C and DIM that are reported in various preclinical investigations. So far, most of the reports about I3C and DIM protective effects against various diseases are only from preclinical studies; this emphasizes the dire need for large-scale clinical trials on these phytochemicals against human diseases. Further, in-depth research is required to improve the bioavailability of these two phytochemicals to achieve the desirable protective effects. Overall, our review emphasizes that I3C and DIM may become potential drug candidates for combating dreadful human diseases.
Collapse
Affiliation(s)
- Yadava Srikanth
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Dontiboina Harikrishna Reddy
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Vinjavarapu Lakshmi Anusha
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Naresh Dumala
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Matte Kasi Viswanadh
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Guntupalli Chakravarthi
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Buchi N. Nalluri
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Ganesh Yadagiri
- Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kakarla Ramakrishna
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| |
Collapse
|
22
|
Li Z, Liu Y, Liu K, Tao X, Hu N, Li W, Duan J. Saponins from Aralia taibaiensis protect against brain ischemia/reperfusion injuries by regulating the apelin/AMPK pathway. Chin J Nat Med 2025; 23:299-310. [PMID: 40122660 DOI: 10.1016/s1875-5364(25)60841-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/07/2024] [Accepted: 05/10/2024] [Indexed: 03/25/2025]
Abstract
Aralia taibaiensi, widely distributed in western China, particularly in the Qinba Mountains, has been utilized as a folk medicine for treating diabetes, gastropathy, rheumatism, and cardiovascular diseases. Saponins from A. taibaiensis (sAT) have demonstrated protective effects against oxidative stress and mitochondrial dysfunction induced by ischemia/reperfusion (I/R). However, the underlying mechanisms remain unclear. In vivo, middle cerebral artery occlusion/reperfusion (MCAO/R) induced inflammatory infiltration, neuronal injury, cell apoptosis, mitochondrial dysfunction, and oxidative stress in the ischaemic penumbra, which were effectively mitigated by sAT. sAT increased the mRNA and protein expression levels of apelin and its receptor apelin/apelin receptors (ARs) both in vivo and in vitro. (Ala13)-Apelin-13 (F13A) and small interfering RNA (siRNA) abolished the regulatory effects of sAT on neuroprotection mediated by adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK)/protein kinase B (Akt). Furthermore, sAT induced apelin/AR expression by simultaneously inhibiting P38 mitogen-activated protein kinase (P38 MAPK)/activating transcription factor 4 (ATF4) and upregulating hypoxia-inducible factor-1α (HIF-1α). Our findings indicate that sAT regulates apelin/AR/AMPK by inhibiting P38 MAPK/ATF4 and upregulating HIF-1a, thereby suppressing oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Zhengrong Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yuwen Liu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Kedi Liu
- TANK Medicinal Biology Institute of Xi'an, Xi'an 710065, China
| | - Xingru Tao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Naping Hu
- Department of Pharmacy, General Hospital of Xinjiang Production and Construction Corps, Urumqi 830092, China
| | - Wangting Li
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jialin Duan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China; Shanghai Minhang Collaborative Innovation Center of Northwestern Polytechnical University, Shanghai 201108, China.
| |
Collapse
|
23
|
Yan X, Quan S, Guo R, Li Z, Bai M, Wang B, Su P, Xu E, Li Y. Calycosin‑7‑O‑β‑D‑glucoside downregulates mitophagy by mitigating mitochondrial fission to protect HT22 cells from oxygen‑glucose deprivation/reperfusion‑induced injury. Mol Med Rep 2025; 31:71. [PMID: 39820475 PMCID: PMC11751592 DOI: 10.3892/mmr.2025.13436] [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: 10/04/2024] [Accepted: 11/26/2024] [Indexed: 01/19/2025] Open
Abstract
Calycosin‑7‑O‑β‑D‑glucoside (CG), a major active ingredient of Astragali Radix, exerts neuroprotective effects against cerebral ischemia; however, whether the effects of CG are associated with mitochondrial protection remains unclear. The present study explored the role of CG in improving mitochondrial function in a HT22 cell model of oxygen‑glucose deprivation/reperfusion (OGD/R). The Cell Counting Kit‑8 assay, flow cytometry, immunofluorescence and western blotting were performed to investigate the effects of CG on mitochondrial function. The results demonstrated that mitochondrial function was restored after treatment with CG, as indicated by reduced mitochondrial reactive oxygen species levels, increased mitochondrial membrane potential and improved mitochondrial morphology. Overactivated mitophagy was revealed to be inhibited by the regulation of proteins involved in fission [phosphorylated‑dynamin‑related protein 1 (Drp1) and Drp1] and mitophagy (LC3, p62 and translocase of outer mitochondrial membrane 20), and mitochondrial biogenesis was demonstrated to be enhanced by increased levels of sirtuin 1 (SIRT1) and peroxisome proliferator‑activated receptor γ coactivator‑1α (PGC‑1α). In addition, neuronal apoptosis was ameliorated by CG, as determined by a decreased rate of apoptosis, and levels of caspase‑3 and Bcl‑2/Bax. In conclusion, the present study demonstrated that CG may alleviate OGD/R‑induced injury by upregulating SIRT1 and PGC‑1α protein expression, and reducing excessive mitochondrial fission and overactivation of mitophagy.
Collapse
Affiliation(s)
- Xiangli Yan
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Siqi Quan
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- College of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Roujia Guo
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- College of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Zibo Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Ming Bai
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Baoying Wang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Pan Su
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Erping Xu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Yucheng Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| |
Collapse
|
24
|
Pu Y, Cheng J, Wang Z, Zhang J, Liang F, Zhang X, Zheng Z, Yin M, Wang Z. Electroacupuncture pretreatment inhibits ferroptosis and inflammation after middle cerebral artery occlusion in rats by activating Nrf2. Histol Histopathol 2025; 40:357-367. [PMID: 38958062 DOI: 10.14670/hh-18-780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
OBJECTIVE Electroacupuncture (EA) pretreatment can effectively increase the tolerance of the brain to ischemic stroke. The mechanism of ischemic tolerance induced by EA is related to Nrf2, but its specific mechanism has not been elucidated. This paper was designed to explore the effect of EA pretreatment on brain injury and the related mechanisms. METHODS Rats were pretreated with EA before middle cerebral artery occlusion (MCAO) modeling. The symptoms of neurological deficit and the volume of cerebral infarction were measured. The levels of inflammatory factors, oxidative stress-related factors, LPO, ROS, and Fe2+ were evaluated by the corresponding kits. Cell apoptosis was determined through TUNEL staining. The mRNA expression of inflammatory factors was examined by RT-qPCR, and the protein expression of ferroptosis-related factors, pyroptosis-related proteins, Keap1, Nrf2, HO-1, and NQO1 by western blotting. RESULTS EA pretreatment improved the symptoms of neurological deficit and reduced the volume of cerebral infarction. EA pretreatment significantly inhibited oxidative stress, inflammatory response, ferroptosis, pyroptosis, and apoptosis in brain tissues of MCAO rats. Mechanistically, EA pretreatment could activate Nrf2 expression and reduce Keap1 expression. CONCLUSION EA pretreatment reduced inflammation and oxidative stress and inhibited ferroptosis by activating Nrf2 expression, ultimately delaying the development of ischemic stroke.
Collapse
Affiliation(s)
- Yanpeng Pu
- Department of Encephalopathy, the Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, PR China
| | - Jingyan Cheng
- Department of Rehabilitation Center, the Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, PR China
| | - Zhenya Wang
- Department of Encephalopathy, the Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, PR China
| | - Jingbo Zhang
- Department of Encephalopathy, the Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, PR China
| | - Fajun Liang
- Department of Encephalopathy, the Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, PR China
| | - Xianbao Zhang
- Department of Encephalopathy, the Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, PR China
| | - Zhijun Zheng
- Department of Encephalopathy, the Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, PR China
| | - Miaomiao Yin
- Department of Encephalopathy, the Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, PR China
| | - Zhen Wang
- Department of Encephalopathy, the Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, PR China.
| |
Collapse
|
25
|
Chen K, Xu B, Long L, Wen H, Zhao Q, Tu X, Wang J, Xu J, Wang H. Inhibition of Phosphodiesterase 4 Suppresses Neuronal Ferroptosis After Cerebral Ischemia/Reperfusion. Mol Neurobiol 2025; 62:3376-3395. [PMID: 39287745 DOI: 10.1007/s12035-024-04495-9] [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: 03/10/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
We have previously shown that inhibition of phosphodiesterase 4 (PDE4) protects against cerebral ischemia/reperfusion injury. However, it remains unclear whether and how PDE4 affects ferroptosis under cerebral ischemia/reperfusion conditions. In this study, we found that overexpression of PDE4B in HT-22 cells exacerbated the detrimental effects of oxygen-glucose deprivation/reoxygenation (OGD/R), including a decrease in cell viability and glutathione (GSH) levels and an increase in Fe2+ content. PDE4B knockdown mitigated the effects of OGD/R, as evidenced by decreased oxidative stress, lactate dehydrogenase (LDH) release, Fe2+ content, and nuclear receptor coactivator 4 (NCOA4) expression. PDE4B knockdown also enhanced the levels of GSH, ferroportin (FPN), and ferritin heavy chain 1 (FTH1). Consistently, inhibition of PDE4 by roflumilast (Roflu) produced similar effects as PDE4B knockdown. Roflu also ameliorated the morphology and membrane potential of the mitochondria. Glutathione peroxidase 4 (GPX4) knockdown blocked the effects of Roflu on cell viability and lipid peroxidation. Moreover, we found that nuclear factor erythroid 2-related factor 2 (Nrf-2) knockdown decreased GPX4 expression. In addition, Nrf-2 knockdown led to enhanced lipid peroxidation, LDH release, and iron levels, while the GSH and FPN levels decreased. More crucially, PDE4 inhibition decreased infarct volume, alleviated oxidative stress, and restored the expression levels of ferroptosis-associated proteins in middle cerebral artery occlusion/reperfusion (MCAO/R) rats. Interestingly, the GPX4 inhibitor RSL3 blocked the neuroprotective effects of Roflu in rats subjected to MCAO/R. Thus, PDE4 inhibition significantly inhibits neuronal ferroptosis by activating the Nrf-2/GPX4 pathway. These data indicate the existence of a novel mechanism underlying the neuroprotective effects of PDE4 inhibition.
Collapse
Affiliation(s)
- Kechun Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bingtian Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lu Long
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Huizhen Wen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qian Zhao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xingxing Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiakang Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiangping Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, 510515, China
| | - Haitao Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
26
|
McDonough A, Weinstein JR. Glial 'omics in ischemia: Acute stroke and chronic cerebral small vessel disease. Glia 2025; 73:495-518. [PMID: 39463002 PMCID: PMC11785505 DOI: 10.1002/glia.24634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
Vascular injury and pathologies underlie common diseases including ischemic stroke and cerebral small vessel disease (CSVD). Prior work has identified a key role for glial cells, including microglia, in the multifaceted and temporally evolving neuroimmune response to both stroke and CSVD. Transcriptional profiling has led to important advances including identification of distinct gene expression signatures in ischemia-exposed, flow cytometrically sorted microglia and more recently single cell RNA sequencing-identified microglial subpopulations or clusters. There is a reassuring degree of overlap in the results from these two distinct methodologies with both identifying a proliferative and a separate type I interferon responsive microglial element. Similar patterns were later seen using multimodal and spatial transcriptomal profiling in ischemia-exposed microglia and astrocytes. Methodological advances including enrichment of specific neuroanatomic/functional regions (such as the neurovascular unit) prior to single cell RNA sequencing has led to identification of novel cellular subtypes and generation of new credible hypotheses as to cellular function based on the enhanced cell sub-type specific gene expression patterns. A ribosomal tagging strategy focusing on the cellular translatome analyses carried out in the acute phases post stroke has revealed distinct inflammation-regulating roles for microglia and astrocytes in this setting. Early spatial transcriptomics experiments using cerebral ischemia models have identified regionally distinct microglial cell clusters in ischemic core versus penumbra. There is great potential for combination of these methods for multi-omics approaches to further elucidate glial responses in the context of both acute ischemic stroke and chronic CSVD.
Collapse
Affiliation(s)
- Ashley McDonough
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington 98195-6465
| | - Jonathan R. Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington 98195-6465
- Department of Neurological Surgery, School of Medicine, University of Washington, Seattle, Washington 98195-6465
| |
Collapse
|
27
|
He L, Lei R, Li S, Zhao X, He X, Yang X, Liu P, Zhang D, Jiang Y. Hirudin promotes cerebral angiogenesis and exerts neuroprotective effects in MCAO/R rats by activating the Wnt/β-catenin pathway. J Stroke Cerebrovasc Dis 2025; 34:108218. [PMID: 39753184 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108218] [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: 09/04/2024] [Revised: 12/17/2024] [Accepted: 12/28/2024] [Indexed: 01/19/2025] Open
Abstract
OBJECTIVE Hirudin has shown potential in promoting angiogenesis and providing neuroprotection in ischemic stroke; however, its therapeutic role in promoting cerebrovascular angiogenesis remains unclear. In this study, we aimed to investigate whether hirudin exerts neuroprotective effects by promoting angiogenesis through the regulation of the Wnt/β-catenin signaling pathway. METHODS An in vitro model of glucose and oxygen deprivation/reperfusion (OGD/R) was established using rat brain microvascular endothelial cells (BMECs). The effects of hirudin on OGD/R cell viability were assessed using the cell counting kit-8 (CCK-8) assay. The angiogenic potential of hirudin was evaluated using Transwell and tube formation assays. In vivo, a middle cerebral artery occlusion/reperfusion (MCAO/R) model was created in rats. The neuroprotective effects of hirudin were assessed using the modified neurological severity score (mNSS), Hematoxylin and eosin (H&E) staining, 2,3,5-Triphenyltetrazolium chloride (TTC) staining, and immunofluorescence staining. Dickkopf-1 (DKK1), a specific inhibitor of this pathway, was introduced in order to investigate the role of the Wnt/β-catenin pathway. The effects of hirudin on the Wnt/β-catenin pathway were examined through immunohistochemistry, western blotting, and reverse transcription quantitative polymerase chain reaction (RT-qPCR). RESULTS Hirudin significantly improved BMEC survival and enhanced both cell migration and tube formation in the OGD/R model. In the MCAO/R model, hirudin reduced the mNSS score, alleviated pathological damage, decreased infarction volume, and increased the expression of key angiogenic factors, including CD34, vascular endothelial growth factor (VEGF), and angiopoietin-2 (Ang-2). In addition, hirudin activated the Wnt/β-catenin pathway, leading to elevated levels of Wnt3a and β-catenin. CONCLUSION Hirudin has substantial neuroprotective effects associated with the promotion of angiogenesis in the ischemic penumbra. This mechanism is mediated by the regulation of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Linrong He
- Department of Gerontology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Ruolan Lei
- Department of Gerontology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Shuangyang Li
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Xiaoying Zhao
- Department of Gerontology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Xinying He
- Department of Gerontology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Xinyue Yang
- Department of Gerontology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Ping Liu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Dechou Zhang
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Yu Jiang
- Department of Gerontology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
28
|
Isaković J, Chin BD, Oberwinter M, Rance HK. From lab coats to clinical trials: Evolution and application of electromagnetic fields for ischemic stroke rehabilitation and monitoring. Brain Res 2025; 1850:149391. [PMID: 39662791 DOI: 10.1016/j.brainres.2024.149391] [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: 08/15/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Stroke is a neurovascular disorder which stands as one of the leading causes of death and disability worldwide, resulting in motor and cognitive impairment. Although the treatment approach depends on the time elapsed, the type of stroke and the availability of care centers, common interventions include thrombectomy or the administration of a tissue plasminogen activator (tPA). While these methods restore blood flow, they fall short in helping patients regain lost function. With that, recent years have seen a rise in novel methods, one of which is the use of electromagnetic fields (EMFs). Due to their ability to impact the charges in their vicinity, thereby altering the immune response and cell signaling, EMFs became suitable candidates for stroke rehabilitation. Based on their characteristics, therapeutic EMFs can be categorized into transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), pulsed (PEMFs) and low frequency (LF-EMFs) electromagnetic fields, among others. In addition to treatment, EMFs are being explored for stroke monitoring, utilizing external EMFs for imaging or recording innate EMFs linked to neural activity. Drawing from research on the effects of EMFs, this review aims to provide a comprehensive overview of the physical principles and molecular mechanisms underlying the action of EMFs, along with a discussion of their application in preclinical studies and clinical trials. Finally, this paper not only addresses the importance of treatment availability and potential side-effects, but also delves into the technical and ethical challenges associated with the use of EMFs, while exploring their prospects and future opportunities.
Collapse
Affiliation(s)
- Jasmina Isaković
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany.
| | - Benjamin Daniel Chin
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany
| | - Moritz Oberwinter
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany
| | - Hannah Katarina Rance
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany
| |
Collapse
|
29
|
Zhang X, Wang SJ, Wan SC, Li X, Chen G. Ozone: complicated effects in central nervous system diseases. Med Gas Res 2025; 15:44-57. [PMID: 39436168 PMCID: PMC11515058 DOI: 10.4103/mgr.medgasres-d-24-00005] [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: 01/15/2024] [Revised: 06/20/2024] [Accepted: 08/25/2024] [Indexed: 10/23/2024] Open
Abstract
Oxidative stress is closely related to various diseases. Ozone can produce redox reactions through its unique response. As a source of the oxidative stress response, the strong oxidizing nature of ozone can cause severe damage to the body. On the other hand, low ozone concentrations can activate various mechanisms to combat oxidative stress and achieve therapeutic effects. Some animal experiments and clinical studies have revealed the potential medical value of ozone, indicating that ozone is not just a toxic gas. By reviewing the mechanism of ozone and its therapeutic value in treating central nervous system diseases (especially ischemic stroke and Alzheimer's disease) and the toxic effects of ozone, we find that ozone inhalation and a lack of antioxidants or excessive exposure lead to harmful impacts. However, with adequate antioxidants, ozone can transmit oxidative stress signals, reduce inflammation, reduce amyloid β peptide levels, and improve tissue oxygenation. Similar mechanisms to those of possible new drugs for treating ischemic stroke and Alzheimer's disease indicate the potential of ozone. Nevertheless, limited research has restricted the application of ozone. More studies are needed to reveal the exact dose-effect relationship and healing effect of ozone.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Shi-Jun Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Si-Cen Wan
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
30
|
Amiranda S, Succoio M, Anzilotti S, Cuomo O, Petrozziello T, Tedeschi V, Finizio A, Mele G, Parkkila S, Annunziato L, De Simone G, Pignataro G, Secondo A, Zambrano N. Pharmacological inhibition of carbonic anhydrases with a positively charged pyridinium sulfonamide phenocopies the neuroprotective effects of Car9 genetic ablation in a murine setting of oxygen/glucose deprivation followed by re-oxygenation and is associated with improved neuronal function in ischemic rats. Heliyon 2025; 11:e42457. [PMID: 40028587 PMCID: PMC11868941 DOI: 10.1016/j.heliyon.2025.e42457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Carbonic anhydrases constitute a family of metalloenzymes vital for maintaining acid-base balance and regulating pH in physio-pathological processes. These findings suggest carbonic anhydrases as potential therapeutic targets for treating pH-associated disorders, including cerebral ischemia, to mitigate hypoxia- and reoxygenation-induced neuronal damage. A focus on carbonic anhydrase IX showed that ischemic stress altered subcellular distributions of this enzyme in rodent neuronal populations. Given the enzyme's canonical membrane localization, we implemented pharmacological inhibition using a membrane-impermeant sulfonamide inhibitor in neuronal models of brain ischemia. The treatments exerted neuroprotective effects on neurons from Car9 knockout mice. Moreover, administration of the sulfonamide inhibitor to rats subjected to transient middle cerebral artery occlusion decreased infarct volumes and improved neurological deficits. Our results support the involvement of carbonic anhydrase IX in postischemic damage and pave the way for possible pharmacological interventions with selective inhibitors in the management of brain ischemia.
Collapse
Affiliation(s)
- Sara Amiranda
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| | - Mariangela Succoio
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| | - Serenella Anzilotti
- Department of Human Sciences and Quality of Life Promotion, Università San Raffaele, Rome, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Tiziana Petrozziello
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Valentina Tedeschi
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Arianna Finizio
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| | - Giorgia Mele
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Ltd, Tampere University Hospital, Tampere, Finland
| | | | - Giuseppina De Simone
- Istituto di Biostrutture e Bioimmagini, CNR, Via Pietro Castellino 111, 80131, Napoli, Italy
| | - Giuseppe Pignataro
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Agnese Secondo
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Nicola Zambrano
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| |
Collapse
|
31
|
Wang GP, Li WJ, Li Y, Ma MX, Guo KK. ADAR1 Promotes NUPR1 A-to-I RNA Editing to Exacerbate Ischemic Brain Injury by Microglia Mediated Neuroinflammation. Neuromolecular Med 2025; 27:16. [PMID: 40009287 DOI: 10.1007/s12017-025-08841-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/19/2025] [Indexed: 02/27/2025]
Abstract
Microglial cells occupy a crucial position as potential therapeutic targets in the context of ischemic stroke (IS). Nonetheless, the intrinsic mechanisms that govern microglial activation in the aftermath of IS remain incompletely elucidated. ADAR1 p150 plays a significant role in immune regulation and stress responses; however, the specific pathways through which it modulates microglial activation and the subsequent mechanisms that unfold following IS have yet to be clearly delineated. The distal middle cerebral artery occlusion (dMCAO) mouse model was utilized to induce IS. The evaluation of infarct volume was conducted through TTC staining, while neurological function was assessed using the modified Neurological Severity Score (mNSS). To evaluate the expression of ADAR1 and apoptosis-related proteins, immunofluorescence and Western blot techniques were employed. BV2 cells were subjected to oxygen-glucose deprivation followed by reperfusion (OGD/R). Additionally, a co-culture system of BV2 cells and neurons was established, and subsequent assessments of neuronal viability and apoptosis were performed using CCK-8 assays and LDH release assays. ADAR1 p150 expression was significantly upregulated in the brains of ischemic mice, particularly within microglial cells. The overexpression of ADAR1 p150 was found to promote microglial activation and enhance pro-inflammatory responses, whereas the knockdown of ADAR1 p150 yielded the opposite effect. Additionally, the knockdown of ADAR1 p150 in microglia resulted in a marked reduction in neuronal apoptosis within the co-culture system. Rescue experiments indicated that the knockdown of NUPR1 partially reinstated the inflammatory response previously induced by ADAR1 p150 knockdown. Notably, ADAR1 p150 knockdown also inhibited A-to-I RNA editing while simultaneously upregulating NUPR1. Furthermore, the reduction of ADAR1 expression was associated with decreased infarct volume, improved neurological outcomes, and a significant attenuation of neuroinflammation in dMCAO mice. ADAR1 p150 enhances the microglial inflammatory response and neuronal apoptosis in IS by facilitating A to I RNA editing of NUPR1.
Collapse
Affiliation(s)
- Guo-Ping Wang
- Department of Anesthesiology, Chang Zhi People's Hospital, 502 Chang Xing Middle Road, Chang Zhi, 046000, Shanxi, China
| | - Wen-Juan Li
- Department of Anesthesiology, Chang Zhi People's Hospital, 502 Chang Xing Middle Road, Chang Zhi, 046000, Shanxi, China
| | - Ye Li
- Department of Pain Medicine, the First Medical Center, Chinese PLA General Hospital, Beijing, 1000853, China
| | - Ming-Xing Ma
- Department of Pain Medicine, the First Medical Center, Chinese PLA General Hospital, Beijing, 1000853, China.
| | - Kai-Kai Guo
- Department of Pain Medicine, the First Medical Center, Chinese PLA General Hospital, Beijing, 1000853, China.
| |
Collapse
|
32
|
Deng Z, Chen X, Zhang R, Kong L, Fang Y, Guo J, Shen B, Zhang L. Delta opioid peptide [D-ala2, D-leu5]-Enkephalin's ability to enhance mitophagy via TRPV4 to relieve ischemia/reperfusion injury in brain microvascular endothelial cells. Stroke Vasc Neurol 2025; 10:32-44. [PMID: 38697767 PMCID: PMC11877439 DOI: 10.1136/svn-2023-003080] [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: 01/03/2024] [Accepted: 04/20/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Local brain tissue can suffer from ischaemia/reperfusion (I/R) injury, which lead to vascular endothelial damage. The peptide δ opioid receptor (δOR) agonist [D-ala2, D-leu5]-Enkephalin (DADLE) can reduce apoptosis caused by acute I/R injury in brain microvascular endothelial cells (BMECs). OBJECTIVE This study aims to explore the mechanism by which DADLE enhances the level of mitophagy in BMECs by upregulating the expression of transient receptor potential vanilloid subtype 4 (TRPV4). METHODS BMECs were extracted and made to undergo oxygen-glucose deprivation/reoxygenation (OGD/R) accompanied by DADLE. RNA-seq analysis revealed that DADLE induced increased TRPV4 expression. The CCK-8 method was used to assess the cellular viability; quantitative PCR (qPCR) was used to determine the mRNA expression of Drp1; western blot was used to determine the expression of TRPV4 and autophagy-related proteins; and calcium imaging was used to detect the calcium influx. Autophagosomes in in the cells' mitochondria were observed by using transmission electron microscopy. ELISA was used to measure ATP content, and a JC-1 fluorescent probe was used to detect mitochondrial membrane potential. RESULTS When compared with the OGD/R group, OGD/R+DADLE group showed significantly enhanced cellular viability; increased expression of TRPV4, Beclin-1, LC3-II/I, PINK1 and Parkin; decreased p62 expression; a marked rise in calcium influx; further increases in mitophagy, an increase in ATP synthesis and an elevation of mitochondrial membrane potential. These protective effects of DADLE can be blocked by a TRPV4 inhibitor HC067047 or RNAi of TRPV4. CONCLUSION DADLE can promote mitophagy in BMECs through TRPV4, improving mitochondrial function and relieving I/R injury.
Collapse
Affiliation(s)
- Zhongfang Deng
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyu Chen
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Ran Zhang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Lingchao Kong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Fang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Jizheng Guo
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Bing Shen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao
| | - Lesha Zhang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
33
|
Lu Y, Li Z, Xu R, Xu Y, Zhang W, Zhang Y, Fang Z, Pan C, Wang X. Impact of fracture fixation surgery on cognitive function and the gut microbiota in mice with a history of stroke. Open Life Sci 2025; 20:20221061. [PMID: 40026365 PMCID: PMC11868713 DOI: 10.1515/biol-2022-1061] [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: 11/05/2024] [Revised: 12/24/2024] [Accepted: 01/12/2025] [Indexed: 03/05/2025] Open
Abstract
Perioperative cognitive dysfunction is a common complication in stroke patients undergoing secondary surgeries. This study investigated the effects of tibial fracture internal fixation (TFIF) surgery on cognitive function and the gut microbiota in mice with a history of stroke. Using the middle cerebral artery occlusion method to induce stroke, we assessed cognitive function via the fear conditioning test and analyzed the gut microbiota through 16S rRNA sequencing. Compared with those in the normal and stroke groups, the cognitive function of the mice in the stroke group that underwent TFIF surgery was significantly impaired. Gut microbiota analysis revealed significant changes in beta diversity, but not in alpha diversity, in these mice. Additionally, TFIF surgery increased microglial activation and IL-1β and lipopolysaccharide (LPS) levels in the brain while reducing α-defensin levels and increasing IL-1β and LPS levels in the colon. These results suggest that TFIF surgery exacerbates cognitive impairment in stroke mice, possibly through alterations in the gut microbiota that impair intestinal defense and promote inflammation. This study highlights the critical role of the gut microbiome in cognitive function and perioperative outcomes, offering insights into potential therapeutic strategies for perioperative cognitive dysfunction in stroke patients.
Collapse
Affiliation(s)
- Yu Lu
- Department of Anesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210029, China
| | - Zixuan Li
- Department of Anesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210029, China
| | - Rukun Xu
- Department of Anesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210029, China
| | - Yajie Xu
- Department of Anesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210029, China
| | - Wenwen Zhang
- Department of Anesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210029, China
| | - Yong Zhang
- Department of Anesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210029, China
| | - Zhaojing Fang
- Department of Anesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210029, China
| | - Cailong Pan
- School of Basic Medical Sciences, Nanjing Medical University, Longmian Avenue 101, Nanjing, 211166, China
| | - Xiaoliang Wang
- Department of Anesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210029, China
| |
Collapse
|
34
|
Gao W, Lv X, Li H, Yan XS, Huo DS, Yang ZJ, Zhang ZG, Jia JX. Dexmedetomidine pretreatment alleviates brain injury in middle cerebral artery occlusion (MCAO) model rats by activating PI3K/AKT/NF-κB signaling pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025:1-11. [PMID: 39995113 DOI: 10.1080/15287394.2025.2469088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) is a prevalent clinical complication associated with reperfusion following ischemic stroke resulting in neuronal damage and cognitive impairment. Dexmedetomidine (DEX), a highly selective α2-adrenoceptor agonist with sedative, and analgesic properties, is frequently utilized as a sedative anesthetic in clinical surgeries, and believed to play a crucial role in the prognosis of patients suffering from CIRI. However, the mechanism underlying DEX in CIRI remains to be determined. This study aimed to investigate the neuroprotective effects of Dex in rats suffering from CIRI. In the treatment group, DEX (50 µg/kg) was administered intraperitoneally 30 min prior to surgery. Middle cerebral artery occlusion (MCAO) used as a model of CIRI occurred with cerebral artery occlusion for 2 h was followed by reperfusion with blood for 24, 72, 120 or 168 h. Neurological function as assessed by the Longa neurological function score test demonstrated significantly reduced neurological scores and increased % infarct size in MCAO group which was blocked by DEX suggesting that DEX might be effective in treating ischemic stroke. In the MCAO animals, 2,3,5-triphenyltetrazolium chloride (TTC) showed large marked areas of cerebral infarction which were diminished in size by DEX. Using Western blot analysis, results showed that in MCAO rats protein expression levels of TNF-α and IL-6 were increased accompanied by reduced protein expression levels of PI3K/AKT signaling pathway. DEX pretreatment reversed the effects of MCAO as evidenced by decrease in protein expression levels of TNF-α and IL-6 associated with elevated protein expression levels of PI3K/AKT/NF-κB signaling pathway. Data demonstrated that DEX pretreatment improved the neuromotor performance and cognitive functions in animals suffering from consequences of MCAO by diminishing inflammation and activation of the PI3K/AKT/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wei Gao
- Department of Anesthesiology, The Third Hospital of Baogang Group, Baotou, China
- Department of Human Anatomy, Baotou Medical College, Baotou, China
| | - Xue Lv
- Department of Human Anatomy, Baotou Medical College, Baotou, China
| | - Hao Li
- Department of Human Anatomy, Baotou Medical College, Baotou, China
| | - Xu-Sheng Yan
- Department of Human Anatomy, Baotou Medical College, Baotou, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| | - Dong-Sheng Huo
- Department of Human Anatomy, Baotou Medical College, Baotou, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| | - Zhan-Jun Yang
- Department of Human Anatomy, Chifeng University, Chi feng, China
| | - Zhi-Guo Zhang
- Department of Anesthesiology, The Third Hospital of Baogang Group, Baotou, China
| | - Jian-Xin Jia
- Department of Human Anatomy, Baotou Medical College, Baotou, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region, Baotou, China
| |
Collapse
|
35
|
Peng Y, Long Y, Wan C. NOD-like receptor X1 promotes autophagy and inactivates NLR family pyrin domain containing 3 inflammasome signaling by binding autophagy-related gene 5 to alleviate cerebral ischemia/reperfusion-induced neuronal injury. J Neuropathol Exp Neurol 2025; 84:223-235. [PMID: 39707156 DOI: 10.1093/jnen/nlae129] [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] [Indexed: 12/23/2024] Open
Abstract
Ischemic strokes pose serious risks to human health. We aimed to elucidate the function of NOD-like receptor X1 (NLRX1) in a rat middle cerebral artery occlusion (MCAO)-induced cerebral ischemia/reperfusion injury (CIRI) model and in an oxygen-glucose deprivation/reperfusion (OGD/R)-treated human microglial cell line (HMC3) model. Following NLRX1 upregulation, infarct volumes were measured with 2,3,5-triphenyltetrazolium chloride staining and pathological examination was conducted with hematoxylin-eosin staining. Results suggested that levels of NLRX1 were decreased in brain tissue of MCAO rats and in OGD/R-stimulated HMC3 cells. NOD-like receptor X1 overexpression mitigated the neuronal damage, reduced tumor necrosis factor-α and interleukin-6 expression, alleviated microglial activation, and induced autophagy in vivo and in vitro. Additionally, a coimmunoprecipitation assay indicated that NLRX1 bound to autophagy-related gene 5 (ATG5) to elevate ATG5 expression in HMC3 cells. Further, the elevated NLR family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a CARD, and cleaved caspase 1 expression in MCAO rats and HMC3 cells with OGD/R induction was reduced after NLRX1 upregulation. Importantly, ATG5 depletion abrogated the effects of NLRX1 elevation on NLRP3 inflammasome signaling. These results indicate that NLRX1 promotes autophagy and inactivates NLRP3 inflammasome signaling by binding ATG5 in experimental cerebral ischemia. These data may help the development of novel therapeutic strategies for ischemic stroke.
Collapse
Affiliation(s)
- Yufen Peng
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Long
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chenyi Wan
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
36
|
Ping Y, Li J, Xie L, Zhao J, Chen X, Chen D, Wang Y, Jiang C, Li X. GPNMB attenuates neuroinflammation and improves ischemic stroke via modulation of PI3K/Akt and p38 MAPK signaling pathways. Brain Res 2025; 1849:149381. [PMID: 39643105 DOI: 10.1016/j.brainres.2024.149381] [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: 10/09/2024] [Revised: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Ischemic stroke is a leading cause of disability and mortality worldwide, with limited effective treatments. Neuroinflammation plays a crucial role in the progression of ischemic brain injury. Glycoprotein nonmetastatic melanoma protein B (GPNMB) has emerged as a potential regulator of inflammation, but its role and underlying mechanisms in ischemic stroke remain largely unknown. METHODS We investigated the expression profile, functional significance, and molecular pathways of GPNMB in ischemic stroke using a mouse model of middle cerebral artery occlusion (MCAO), transcriptome sequencing, and human serum samples. The effects of GPNMB knockdown on stroke outcomes, neuroinflammation, and neuronal damage were assessed in vivo. Bioinformatic analyses and experimental validation were performed to identify the downstream signaling pathways of GPNMB. RESULTS GPNMB was highly upregulated in the ischemic brain, with its expression peaking at 3-7 days post-MCAO. Serum GPNMB levels were elevated in ischemic stroke patients and correlated with stroke severity. GPNMB knockdown exacerbated stroke outcomes, neuroinflammation, and neuronal damage. Mechanistically, GPNMB positively modulated the PI3K/Akt/GSK3β pathway while negatively regulating p38 MAPK, JNK, and ERK activation. GPNMB knockdown enhanced the expression of NF-κB, a master transcriptional regulator of inflammation. CONCLUSION GPNMB is highly upregulated in the ischemic brain and confers neuroprotection against ischemic injury by modulating neuroinflammation via the PI3K/Akt and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Yukun Ping
- Clinical Medical College, Yangzhou University, Yangzhou 225009, China; Northern Jiangsu People's Hospital Affliated to Yangzhou University, Yangzhou 225001, China
| | - Jiyu Li
- Department of Orthopedic Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Linlin Xie
- Clinical Medical College, Yangzhou University, Yangzhou 225009, China; Northern Jiangsu People's Hospital Affliated to Yangzhou University, Yangzhou 225001, China
| | - Jie Zhao
- Clinical Medical College, Yangzhou University, Yangzhou 225009, China; Northern Jiangsu People's Hospital Affliated to Yangzhou University, Yangzhou 225001, China
| | - Xuyu Chen
- Clinical Medical College, Yangzhou University, Yangzhou 225009, China
| | - Danni Chen
- Northern Jiangsu People's Hospital Affliated to Yangzhou University, Yangzhou 225001, China
| | - Yamin Wang
- Sanquan College of Xinxiang Medical University, Xinxiang 453003, China
| | - Chao Jiang
- Northern Jiangsu People's Hospital Affliated to Yangzhou University, Yangzhou 225001, China.
| | - Xiaobo Li
- Northern Jiangsu People's Hospital Affliated to Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
37
|
ElBassiouny A, Shehata MSA, Zaki AS, Bedros RY, El-Sudany AH, Nasser AA. Cerebrolysin as an adjuvant therapy after mechanical thrombectomy in large vessel occlusion cardioembolic stroke: a propensity score matching analysis. Front Neurol 2025; 16:1510284. [PMID: 40027163 PMCID: PMC11868704 DOI: 10.3389/fneur.2025.1510284] [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: 10/12/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Endovascular recanalization therapy has demonstrated considerable efficacy in the treatment of acute ischemic stroke (AIS). However, not all patients appear to benefit on the long term from this therapy. No studies have assessed the role of Cerebrolysin following mechanical thrombectomy (MT). The present study was conducted to evaluate the safety and efficacy of Cerebrolysin as add-on treatment to MT in patients with cardioembolic AIS. Methods This study evaluated 150 patients admitted to the stroke unit. Data were prospectively collected from 75 patients with cardioembolic AIS and National Institutes of Health Stroke Scale (NIHSS) ≥10, who underwent successful MT ± recombinant tissue plasminogen activator (rt-PA). Patients fulfilling inclusion criteria were consecutively enrolled and treated with Cerebrolysin at a daily dose of 30 ml for 14 days, with treatment initiated within 8 h following MT. Patients were compared with a historical control group of 75 well-matched patients who underwent MT ± rt-PA but did not receive Cerebrolysin. The primary outcome measure was a favorable modified Rankin Scale (mRS = 0-2) at day 90. Secondary parameters included the NIHSS, the Montreal Cognitive Assessment (MoCA), the rate of hemorrhagic transformation, mortality, and adverse events. Propensity score matching was performed to match the variables between the compared groups. Results and discussion The overall results demonstrated that patients treated with Cerebrolysin exhibited a significantly higher proportion of mRS scores of 0-2 at day 90 (64% vs. 34.7%) in comparison to the control group. This finding was consistent with lower NIHSS and mRS scores at all study visits, and a lower any hemorrhagic transformation rate (20% vs. 57.3%). Furthermore, the logistic regression analysis revealed that patients with favorable mRS scores were less likely to undergo hemorrhagic transformation (odds ratio = 2.75, 95% confidence interval = 1.17, 6.45; p = 0.002). The administration of Cerebrolysin as an add-on treatment resulted in a significant benefit for AIS patients following MT, characterized by an improvement in mRS and NIHSS scores, along with a reduced rate of hemorrhagic transformation. The administration of Cerebrolysin was safe and well tolerated. Further studies are required to confirm these results.
Collapse
Affiliation(s)
- Ahmed ElBassiouny
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed S. A. Shehata
- Faculty of Medicine, Zagazig University, Zagazig, Egypt
- Egyptian Fellowship of Neurology, Ministry of Health, Cairo, Egypt
| | - Amr S. Zaki
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Rady Y. Bedros
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Azza Abdel Nasser
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
38
|
Pan J, Chai X, Li C, Wu Y, Ma Y, Wang S, Xue Y, Zhao Y, Chen S, Zhu X, Zhao S. Eucommia ulmoides Oliv. Bark Extracts Alleviate MCAO/Reperfusion-Induced Neurological Dysfunction by Suppressing Microglial Inflammation in the Gray Matter. Int J Mol Sci 2025; 26:1572. [PMID: 40004043 PMCID: PMC11855810 DOI: 10.3390/ijms26041572] [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: 12/24/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Ischemic stroke ranks as the second leading cause of global mortality. The limited time for effective thrombolytic treatment has prompted the exploration of alternative prevention approaches. Eucommia ulmoides (E. ulmoides) Oliv. bark has shown multiple pharmacological effects, including neuroprotection, anti-inflammation and autophagy modulation. This study aims to elucidate the neuroprotective effects of water extract of E. ulmoides (WEU) supplementation in a middle cerebral artery occlusion (MCAO) mouse model and to further explore the underlying molecular mechanisms. Seven bioactive compounds in WEU-aucubin, chlorogenic acid, geniposidic acid, quercetin, protocatechuic acid, betulin and pinoresinol diglucoside-were identified using HPLC-MS. Our results showed that WEU supplementation significantly decreased infarct volume and ameliorated neurological dysfunction in mice following MCAO/reperfusion (MCAO/R) injury. Furthermore, the administration of WEU significantly attenuated microglia activation induced by cortical ischemia in mice and inhibited the production of pro-inflammatory mediators, including interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Importantly, in contrast with the vehicle group, the protein expression levels of Toll-like receptor 4 (TLR4), phospho-p38 (p-p38) and nuclear factor kappa B (NF-κB) were reduced in the WEU group. Therefore, this present study provides evidence that E. ulmoides improves neurological behaviors by suppressing neuroinflammation and inhibiting the activation of the TLR4/ p38 MAPK and NF-κB pathways in mice after ischemia, which indicates that E.ulmoides is a promising candidate for alleviating gray matter ischemic change.
Collapse
Affiliation(s)
- Jiarong Pan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Xuejun Chai
- College of Basic Medicine, Xi’an Medical University, Xi’an 710021, China;
| | - Cixia Li
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China;
| | - Yongji Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Yue Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Songlin Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Yuhuan Xue
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Yongkang Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Shulin Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (J.P.); (Y.W.); (Y.M.); (S.W.); (Y.X.); (Y.Z.); (S.C.)
| |
Collapse
|
39
|
Basnet N, Cho H, Sapkota A, Park S, Lim C, Gaire BP, Kim D, Lee JY, Been JH, Lee S, Lee BY, Choi JW, Kim S. Blocking S1P 4 signaling attenuates brain injury in mice with ischemic stroke. J Adv Res 2025:S2090-1232(25)00110-9. [PMID: 39952320 DOI: 10.1016/j.jare.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/17/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025] Open
Abstract
INTRODUCTION The functions of S1P receptors have been revealed using genetic and pharmacological tools, including the potent non-selective modulator FTY720. However, studies on subtype-specific agonists and antagonists are limited; hence, the role of S1P4 remains unclear. OBJECTIVES To identify a novel function of S1P4 as a pathogenic factor in stroke using a newly developed S1P4-selective modulator and S1P4 knockdown. METHODS Heteroaromatic analogs of FTY720 were synthesized, a β-arrestin assay was conducted against S1P receptors, and the developed compound (NXC736) was characterized as a functional S1P4 antagonist. To clarify the function of S1P4, the therapeutic potential of NXC736 in ischemic stroke was determined using a transient middle cerebral artery occlusion (tMCAO) mouse model, which was validated using S1P4 knockdown. The S1P4-dependent pathogenic mechanisms were determined using immunohistochemical and biochemical analyses. RESULTS Molecular modeling studies provide valuable clues for understanding S1P4 selectivity of NXC736. NXC736 contains a triazole ring instead of a phenyl ring and exhibits S1P4-selective activity as a functional antagonist. Its action on S1P4 does not require phosphorylation by sphingosine kinase 2. Notably, NXC736 exhibited substantial therapeutic activity against ischemic stroke by attenuating tMCAO-induced acute brain injuries, including brain infarction, neurological deficits, and neuronal apoptosis. This suggested that S1P4 is a pathogenic factor in ischemic stroke. This function was confirmed using AAV-based S1P4 knockdown. NXC736 or S1P4 knockdown attenuated blood-brain barrier disruption, neutrophil infiltration, microglial activation and proliferation, and the upregulation of pro-inflammatory cytokines, thereby demonstrating that S1P4 influences neuroinflammatory responses in ischemic stroke. The underlying mechanisms were activation of NLRP3 inflammasome, NF-κB, and MAPKs. S1P4 also contributed to chronic brain injuries caused by ischemic stroke because NXC736 exerted long-term neuroprotective effects against tMCAO challenge. CONCLUSION Using a functional S1P4 antagonist (NXC736) and a genetic tool for S1P4 knockdown, we identified S1P4 as a novel pathogenic factor in ischemic stroke.
Collapse
Affiliation(s)
- Nikita Basnet
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon 21936, Korea
| | - Hyunkyung Cho
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Arjun Sapkota
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon 21936, Korea
| | - Seungbae Park
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Chaemin Lim
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Bhakta Prasad Gaire
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon 21936, Korea
| | - Donghee Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon 21936, Korea
| | - Joo-Youn Lee
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Jae Hui Been
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Seunghee Lee
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Bong Yong Lee
- Nextgen Bioscience, 228-17 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi-do 13487, Korea
| | - Ji Woong Choi
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Sanghee Kim
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| |
Collapse
|
40
|
Zhang S, Li R, Song M, Han J, Fan X. Exploration of M2 macrophage membrane as a biotherapeutic agent and strong synergistic therapeutic effects in ischemic stroke. J Control Release 2025; 378:476-489. [PMID: 39561947 DOI: 10.1016/j.jconrel.2024.11.033] [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: 07/18/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
The macrophage-derived membrane is widely applied in the targeting nanocarrier for its inflammatory tendency and long circulation ability due to the preservation of membrane protein. Few studies reported the application of macrophage membranes as biotherapeutic agents. To verify the ability of macrophage membrane as a biotherapeutic agent, ischemic stroke was selected as the model disease. Inspired by the features of macrophages infiltrating the ischemic core and the talent of M2 macrophages in modulating the inflammatory microenvironment, an M2 macrophage membrane (M2M)-disguised poly lactic-co-glycolic acid nanoparticles loaded with baicalin (BA) (M2M@BANPs) is developed. The results in vivo and in vitro indicate that M2M@BANPs could efficiently and actively target ischemic brain tissue and accumulate in microglia and neurons due to the coating of M2M. Furthermore, M2M and M2M@BANPs exhibit significant therapeutic effects in salvaging brain tissue damage and neurological functional recovery by reprogramming microglia from M1 to M2, reducing neutrophil infiltration and inhibiting neuronal apoptosis. Together, our fabrication provides a new insight and an applicative perspective for M2M in the therapy of ischemic stroke.
Collapse
Affiliation(s)
- Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Meiying Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jin Han
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
41
|
Li Y, Ding S, Wang Y. Targeting the cholinergic anti-inflammatory pathway: an innovative strategy for treating diseases. Mol Biol Rep 2025; 52:199. [PMID: 39903351 DOI: 10.1007/s11033-025-10288-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
The cholinergic anti-inflammatory pathway (CAP) is comprised of the vagus nerve, acetylcholine, nicotinic acetylcholine receptors, the spleen, and the splenic nerve. It represents a sophisticated neuroimmune axis that critically regulates the crosstalk between the nervous system and the immune response via the vagus nerve. Here, we provided a nuanced exploration of the CAP's role in curbing inflammatory processes and its broad therapeutic potential across a spectrum of diseases. We meticulously dissect the intricate mechanisms by which the CAP modulates key signaling cascades, including the NF-κB, JAK2/STAT3, MAPK/ERK, PI3K/AKT, COX2/PGE2, and NRF2/HO-1 pathways, which are quintessential in the pathogenesis of various conditions. Additionally, we also summarized the CAP's profound implications in the management of inflammatory diseases, neurodegenerative disorders, metabolic syndromes, and oncological malignancies, elucidating its capacity to mitigate disease severity and progression through sophisticated immune modulation. The modulation of the CAP is suggested as a novel strategy that could potentially transform treatment approaches for a variety of conditions. However, the precise cellular and molecular underpinnings of the CAP's effects, as well as its translatability to clinical settings, remain subjects of ongoing investigation. The review calls for further research to demystify the mechanisms of the CAP and to harness its therapeutic potential fully, with the aim of developing innovative and efficacious treatment modalities that exploit the pathway's unique attributes.
Collapse
Affiliation(s)
- Yifan Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- School of Medicine, Hangzhou Normal University, Hangzhou, 311121, China
| | - Shufan Ding
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yongjie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
42
|
Wang C, Li S, Li Q, Xi H, Li J, Zhu Q, Wu P, Zhu Y, Mao Y. H 2S Donor SPRC Ameliorates Ischemic Stroke by Upregulating CD24. CNS Neurosci Ther 2025; 31:e70243. [PMID: 39953809 PMCID: PMC11829115 DOI: 10.1111/cns.70243] [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: 08/11/2024] [Revised: 12/10/2024] [Accepted: 12/24/2024] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Ischemic stroke is well-known for its high mortality and morbidity, but its treatment remains to be explored due to the current limitations. For example, severe neuroinflammation occurs after ischemic stroke; however, effective neuroinflammatory inhibitors are still lacking. Thus, the development of new therapeutic targets of inhibiting neuroinflammation is urgent. CD24 is a small heavy glycosylated protein, which plays a critical role in neural development and acts as an inflammatory suppressor in tumors and autoimmune diseases. But the role of CD24 in ischemic stroke remains unknown. AIMS The role of CD24 in ischemic stroke should be explored. Additionally, the potential relationship between the H2S donor, S-propargyl-cysteine (SPRC) and CD24 in ischemic stroke should be revealed. METHODS Mechanism studies have been performed both in vitro and in vivo to verify the CD24-mediated inflammation and migration. SPRC has been applied to treat ischemic stroke, and its potential association with CD24 has been studied. RESULTS The overexpression of CD24 can inhibit the nuclear factor kappa B (NF-κB) inflammatory signaling pathway and promote the migration ability of M2 microglia cells via Src/Fak/Pyk2 signaling pathway in an inflammatory model of BV2 cells. SPRC can upregulate the level of endogenous H2S via cystathionase-β-synthase (CBS) and it indirectly plays a role in upregulating CD24. CONCLUSIONS CD24 could be a potential target of inhibiting neuroinflammation. SPRC reduces inflammation in ischemic stroke by regulating the CD24/Iκ-Bα/NF-κB inflammatory signaling pathway and improves the migration ability of M2 microglia via CD24/Src/Fak/Pyk2 signaling pathway, which further alleviates the inflammatory response at the lesion.
Collapse
Affiliation(s)
- Chenye Wang
- Department of Pharmacology, the Key Laboratory of Smart Drug Delivery (Ministry of Education), School of PharmacyMinhang HospitalFudan UniversityShanghaiChina
| | - Sha Li
- Department of Pharmacology, the Key Laboratory of Smart Drug Delivery (Ministry of Education), School of PharmacyMinhang HospitalFudan UniversityShanghaiChina
| | - Qixiu Li
- Department of Pharmacology, the Key Laboratory of Smart Drug Delivery (Ministry of Education), School of PharmacyMinhang HospitalFudan UniversityShanghaiChina
| | - Haiyan Xi
- Department of Pharmacology, the Key Laboratory of Smart Drug Delivery (Ministry of Education), School of PharmacyMinhang HospitalFudan UniversityShanghaiChina
| | - Jiejia Li
- School of Pharmacy and Laboratory of Drug Discovery from Natural Resources and IndustrializtionMacau University of Science and TechnologyMacauChina
- School of PharmacyProvincial Key Laboratory of Inflammation and Molecular Drug TargetInstitute for Translational NeuroscienceAffiliated Hospital 2 of Nantong UniversityCentre for Neural Developmental and Degenerative ResearchNantong UniversityNantongChina
| | - Qing Zhu
- School of PharmacyProvincial Key Laboratory of Inflammation and Molecular Drug TargetInstitute for Translational NeuroscienceAffiliated Hospital 2 of Nantong UniversityCentre for Neural Developmental and Degenerative ResearchNantong UniversityNantongChina
| | - Pinwen Wu
- Department of Pharmacology, the Key Laboratory of Smart Drug Delivery (Ministry of Education), School of PharmacyMinhang HospitalFudan UniversityShanghaiChina
| | - Yi‐Zhun Zhu
- Department of Pharmacology, the Key Laboratory of Smart Drug Delivery (Ministry of Education), School of PharmacyMinhang HospitalFudan UniversityShanghaiChina
- School of Pharmacy and Laboratory of Drug Discovery from Natural Resources and IndustrializtionMacau University of Science and TechnologyMacauChina
| | - Yicheng Mao
- Department of Pharmacology, the Key Laboratory of Smart Drug Delivery (Ministry of Education), School of PharmacyMinhang HospitalFudan UniversityShanghaiChina
| |
Collapse
|
43
|
Lu Y, Shi M, Huang W, Li F, Liang H, Liu W, Huang T, Xu Z. Diosmin alleviates NLRP3 inflammasome-dependent cellular pyroptosis after stroke through RSK2/CREB pathway. Brain Res 2025; 1848:149336. [PMID: 39547499 DOI: 10.1016/j.brainres.2024.149336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
In the context of our previous analyses on the main active ingredients of Jieyudan, a classic formula targeting aphasia in stroke, we further delve into the function and mechanisms of its active ingredient, Diosmin (DM), which may exert neuroprotective effects, in ischemic stroke. Herein, bioinformatics analysis revealed targets of DM and their intersection with differentially expressed genes in ischemic stroke. Middle cerebral artery occlusion (MCAO) rats and oxygen-glucose deprivation (OGD) cells were used to construct in vivo and in vitro models of ischemic stroke. The effects of DM on MCAO rats were assessed by Zea-Longa score, Morris water maze, TTC staining, Nissl staining, immunohistochemistry, and Western blot. At the cellular level, cell counting kit-8 assay and Western blot were carried out to verify the mechanism of DM in ischemic stroke. In vivo, DM decreased neurological deficit score, cerebral infarct volume and neuronal damage, and improved cognitive function in MCAO rats. In vitro, DM increased the viability of OGD-treated cells. In addition, DM down-regulated the expressions of NLR family pyrin domain containing 3 (NLRP3) and pyroptosis-associated proteins, while up-regulating ribosomal protein S6 kinase A3 (RSK2) level and activating cyclic-AMP response element-binding protein (CREB) signaling. Conversely, RSK2 inhibitor LJH685 reduced the viability and promoted pyroptosis-associated protein levels, which also partially reversed the effects of DM in vitro. Collectively, DM plays a therapeutic role in ischemic stroke by inhibiting NLRP3 inflammasome-mediated cellular pyroptosis via the RSK2/CREB pathway.
Collapse
Affiliation(s)
- Yanfei Lu
- Department of Pharmacy, Zhejiang Rehabilitation Medical Center (Rehabilitation Hospital Affiliated to Zhejiang Chinese Medical University), China
| | - Min Shi
- Department of Pharmacy, Zhejiang Rehabilitation Medical Center (Rehabilitation Hospital Affiliated to Zhejiang Chinese Medical University), China
| | - Wei Huang
- Department of Pharmacy, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincal Hospital of Traditional Chinese Medicine), China
| | - Fenfen Li
- College of Pharmacy, Zhejiang Chinese Medical University, China
| | - Haowei Liang
- Graduate School of Zhejiang Chinese Medical University, China
| | - Wenbing Liu
- Department of Cardiopulmonary Rehabilitation, the Third Affiliated Hospital of Zhejiang Chinese Medical University, China
| | - Tianyi Huang
- Department of Pharmacy, Zhejiang Rehabilitation Medical Center (Rehabilitation Hospital Affiliated to Zhejiang Chinese Medical University), China
| | - Zhen Xu
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, China.
| |
Collapse
|
44
|
Guo W, Hu C, Wang Y, Zhang W, Zhang S, Peng J, Wang Y, Wu J. NO-releasing double-crosslinked responsive hydrogels accelerate the treatment and repair of ischemic stroke. Acta Pharm Sin B 2025; 15:1112-1125. [PMID: 40177574 PMCID: PMC11959942 DOI: 10.1016/j.apsb.2025.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/28/2024] [Accepted: 12/07/2024] [Indexed: 04/05/2025] Open
Abstract
Stroke is a global disease that seriously threatens human life. The pathological mechanisms of ischemic stroke include neuroinflammation, oxidative stress, and the destruction of blood vessels at the lesion site. Here, a biocompatible in situ hydrogel platform was designed to target multiple pathogenic mechanisms post-stroke, including anti-inflammation, anti-oxidant, and promotion of angiogenesis. Double-crosslinked responsive multifunctional hydrogels could quickly respond to the pathological microenvironment of the ischemic damage site and mediate the delivery of nitric oxide (NO) and ISO-1 (inhibitor of macrophage migration inhibitory factor, MIF). The hydrogel demonstrated good biocompatibility and could scavenge reactive oxygen species (ROS) and inflammatory cytokines, such as interleukin-6 (IL-6), interleukin-10 (IL-10), and MIF. In a mouse stroke model, hydrogels, when situated within the microenvironment of cerebral infarction characterized by weak acidity and elevated ROS release, would release anti-inflammatory nanoparticles rapidly that exert an anti-inflammatory effect. Concurrently, NO was sustained release to facilitate angiogenesis and provide neuroprotective effects. Neurological function was significantly improved in treated mice as assessed by the modified neurological severity score, rotarod test, and open field test. These findings indicate that the designed hydrogel held promise for sustained delivery of NO and ISO-1 to alleviate cerebral ischemic injury by responding to the brain's pathological microenvironment.
Collapse
Affiliation(s)
- Wen Guo
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610064, China
| | - Yue Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610064, China
| | - Wen Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610064, China
| | - Shaomin Zhang
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Peng
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Med-X Center for Materials, Sichuan University, Chengdu 610064, China
| | - Jinhui Wu
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
45
|
Li Y, Liu L, Yang Z, Li M, Tang T, Xu J. The association between dietary fiber intake and all-cause mortality and cardiovascular disease mortality in patients with stroke: a retrospective cohort study of NHANES. Nutr Res Pract 2025; 19:41-54. [PMID: 39959748 PMCID: PMC11821774 DOI: 10.4162/nrp.2025.19.1.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/16/2024] [Accepted: 09/05/2024] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND/OBJECTIVES Stroke represents the primary cause of death and persistent disability globally, leading to around 5.5 million annual patient fatalities. The objective was to explore the relationship of dietary fiber with all-cause and cardiovascular disease (CVD) mortality risk in patients with stroke. SUBJECTS/METHODS We extracted stroke patients' data from the National Health and Nutrition Examination Survey (NHANES) database. All-cause and CVD mortality were outcomes. Dietary fiber consists of non-digestible forms of carbohydrates, usually polysaccharides that originate from plant-based foods. Covariates including demographic data, vital signs, comorbidities, laboratory parameters, and medication use were screened using the weighted multivariate Cox regression models with backward elimination. Weighted univariate and multivariate Cox regression models were performed to explore the relationship between dietary fiber intake and all-cause/CVD mortality, with hazard ratios (HRs) and 95% confidence intervals (CIs). The association was further investigated in different subgroups. RESULTS A total of 1,578 patients with stroke were included, of whom 688 (43.6%) died. Total fiber and vegetable fiber intake were analyzed as categorical variables, and the lowest intake was considered reference groups. High intake of total fiber (HR, 0.73; 95% CI, 0.57-0.94) and high intake of vegetable fiber (HR, 0.63; 95% CI, 0.48-0.82) were related to lower all-cause mortality risk in individuals with stroke. Similar findings were also observed between higher total fiber (HR, 0.56; 95% CI, 0.37-0.85) and vegetable fiber intake (HR, 0.57; 95% CI, 0.36-0.89) with decreased CVD mortality risk. The relationship between higher total fiber intake and lower all-cause mortality risk was discovered in individuals aged ≥ 60 yrs, smoking, non-CVD, and chronic kidney disease (CKD). High total fiber, or vegetable fiber consumption was linked to lower CVD mortality risk in stroke individuals aged ≥ 60 yrs, females, body mass index ≥ 30 kg/m2, non-smoking, and CKD. CONCLUSION Dietary fiber intake and vegetable fiber intake may benefit the prognosis of patients with stroke. Increasing dietary fiber consumption, especially vegetable fiber intake, potentially benefits the prognosis of stroke patients.
Collapse
Affiliation(s)
- Yanli Li
- Department of Traditional Chinese Medicine, Beijing Boai Hospital, China Rehabilitation Research Center, School of Rehabilitation Medicine, Capital Medical University, Beijing 100068, P.R. China
| | - Lanqun Liu
- Department of Traditional Chinese Medicine, Beijing Boai Hospital, China Rehabilitation Research Center, School of Rehabilitation Medicine, Capital Medical University, Beijing 100068, P.R. China
| | - Zufu Yang
- Department of Traditional Chinese Medicine, Beijing Boai Hospital, China Rehabilitation Research Center, School of Rehabilitation Medicine, Capital Medical University, Beijing 100068, P.R. China
| | - Mingyu Li
- Department of Traditional Chinese Medicine, Beijing Boai Hospital, China Rehabilitation Research Center, Beijing 100068, P.R. China
| | - Tao Tang
- Department of Traditional Chinese Medicine, Beijing Boai Hospital, China Rehabilitation Research Center, Beijing 100068, P.R. China
| | - Jimin Xu
- Department of Traditional Chinese Medicine, Beijing Boai Hospital, China Rehabilitation Research Center, School of Rehabilitation Medicine, Capital Medical University, Beijing 100068, P.R. China
| |
Collapse
|
46
|
Yin X, Zhang C. Circulating miR-574-5p shows diagnostic and prognostic significance and regulates oxygen-glucose deprivation (OGD)-induced inflammatory activation of microglia by targeting ATP2B2. Mol Cell Probes 2025; 79:102016. [PMID: 39880338 DOI: 10.1016/j.mcp.2025.102016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/14/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Early screening is critical for the prevention of ischemic stroke. miR-574-5p was considered a promising biomarker for ischemic stroke but lacks direct confirmation. This study evaluated miR-574-5p in discriminating ischemic stroke and predicting the severity and prognosis of patients, aiming to provide novel insights into the clinical prevention of ischemic stroke. METHODS The clinical significance of miR-574-5p was evaluated in 103 ischemic stroke patients with 87 healthy individuals as control. The potential of serum miR-574-5p in the diagnosis and prognosis of ischemic stroke was assessed by ROC and logistic regression analyses. In vitro, oxygen-glucose deprivation (OGD)-induced microglia was established. The regulation of inflammation, oxidative stress, and proliferation of microglia by miR-574-5p were assessed by cell transfection. The downstream targets of miR-574-5p were predicted from public databases, and the targeting relationship was evaluated by luciferase reporter assay. RESULTS Reducing serum miR-574-5p was observed in ischemic stroke patients relative to healthy individuals, which discriminated ischemic stroke patients. Serum miR-574-5p was negatively correlated with the NIHSS score of ischemic stroke patients and was identified as a risk factor for patients' adverse prognosis. In OGD-induced microglia, overexpressing miR-574-5p could alleviate OGD-induced inflammation and oxidative stress and promote cell growth. Among predicted targets, ATP2B2 was upregulated in ischemic stroke and showed a negative correlation with miR-574-5p. miR-574-5p negatively regulated ATP2B2 in OGD-induced microglia, and the overexpression of ATP2B2 reversed the protective effect of miR-574-5p. CONCLUSION miR-574-5p acted as a biomarker for ischemic stroke and mediated neuroinflammation via targeting ATP2B2.
Collapse
Affiliation(s)
- Xia Yin
- Department of Rehabilitation Medicine, Weifang No.2 People's Hospital, Weifang, 261042, China
| | - Chunlei Zhang
- Department of Neonatology, Weifang Maternal and Child Health Hospital, Weifang, 261042, China.
| |
Collapse
|
47
|
Liu P, Liu X, Ren M, Liu X, Shi X, Li M, Li S, Yang Y, Wang D, Wu Y, Yin F, Guo Y, Yang R, Cheng M, Xin Y, Kang J, Huang B, Ren K. Neuronal cathepsin S increases neuroinflammation and causes cognitive decline via CX3CL1-CX3CR1 axis and JAK2-STAT3 pathway in aging and Alzheimer's disease. Aging Cell 2025; 24:e14393. [PMID: 39453382 PMCID: PMC11822647 DOI: 10.1111/acel.14393] [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: 04/07/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Aging is an intricate process involving interactions among multiple factors, which is one of the main risks for chronic diseases, including Alzheimer's disease (AD). As a member of cysteine protease, cathepsin S (CTSS) has been implicated in inflammation across various diseases. Here, we investigated the role of neuronal CTSS in aging and AD started by examining CTSS expression in hippocampus neurons of aging mice and identified a significant increase, which was negatively correlated with recognition abilities. Concurrently, we observed an elevation of CTSS concentration in the serum of elderly people. Transcriptome and fluorescence-activated cell sorting (FACS) results revealed that CTSS overexpression in neurons aggravated brain inflammatory milieu with microglia activation to M1 pro-inflammatory phenotype, activation of chemokine C-X3-C-motif ligand 1 (CX3CL1)-chemokine C-X3-C-motif receptor 1 (CX3CR1) axis and janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) pathway. As CX3CL1 is secreted by neurons and acts on the CX3CR1 in microglia, our results revealed for the first time the role of neuron CTSS in neuron-microglia "crosstalk." Besides, we observed elevated CTSS expression in multiple brain regions of AD patients, including the hippocampus. Utilizing CTSS selective inhibitor, LY3000328, rescued AD-related pathological features in APP/PS1 mice. We further noticed that neuronal CTSS overexpression increased cathepsin B (CTSB) activity, but decreased cathepsin L (CTSL) activity in microglia. Overall, we provide evidence that CTSS can be used as an aging biomarker and plays regulatory roles through modulating neuroinflammation and recognition in aging and AD process.
Collapse
Affiliation(s)
- Pei‐Pei Liu
- Clinical Systems Biology LaboratoriesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiao‐Hui Liu
- Clinical Systems Biology LaboratoriesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Ming‐Jing Ren
- Department of NephropathyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiao‐Tong Liu
- Department of Clinical LaboratoryThe First Hospital of Yongnian DistrictHebeiChina
| | - Xiao‐Qing Shi
- Department of Clinical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Ming‐Li Li
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shu‐Ang Li
- Clinical Systems Biology LaboratoriesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yang Yang
- Clinical Systems Biology LaboratoriesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Dian‐Dian Wang
- Clinical Systems Biology LaboratoriesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yue Wu
- Clinical Systems Biology LaboratoriesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Fan‐Xiang Yin
- Translational Medical CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yan‐Hong Guo
- Department of NephropathyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Run‐Zhou Yang
- Clinical Systems Biology LaboratoriesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Meng Cheng
- Henan BranchBank of ChinaZhengzhouHenanChina
| | - Yong‐Juan Xin
- Department of Child and Adolescent HealthPrecision Nutrition Innovation Center, School of Public Health, Zhengzhou UniversityZhengzhouHenanChina
| | - Jian‐Sheng Kang
- Clinical Systems Biology LaboratoriesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Bing Huang
- Pain and Related Disease Research LaboratoryShantou University Medical CollegeShantouGuangdongChina
| | - Kai‐Di Ren
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
48
|
Sun K, Shi R, Yu X, Wang Y, Zhang W, Yang X, Zhang M, Wang J, Jiang S, Li H, Kang B, Li T, Zhao S, Ai Y, Qiu J, Wang H, Wang X. Noninvasive imaging biomarker reveals invisible microscopic variation in acute ischaemic stroke (≤ 24 h): a multicentre retrospective study. Sci Rep 2025; 15:3743. [PMID: 39885213 PMCID: PMC11782523 DOI: 10.1038/s41598-025-88016-1] [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: 08/29/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
To develop and validate non-contrast computed tomography (NCCT)-based radiomics method combines machine learning (ML) to investigate invisible microscopic acute ischaemic stroke (AIS) lesions. We retrospectively analyzed 1122 patients from August 2015 to July 2022, whose were later confirmed AIS by diffusion-weighted imaging (DWI). However, receiving a negative result was reported by radiologists according to the NCCT images. Patients in five institutions (n = 592) were combined to generate training and internal validation sets, remaining in three institutions as external validation sets (n = 204, 53 and 273). Through a series of procedures: head alignment, co-registration of NCCT and DWI, the volume of interest delineation and feature extraction. Multiple ML models (random forest, RF; support vector machine, SVM; logistic regression, LR; multilayer perceptron, MLP) were used to discriminate microscopic AIS and non-AIS. Among 1122 patients included (760 men [67.7%]; median [range] age, 64 [21-96] years). After least absolute shrinkage and selection operator (LASSO) algorithm, 44 optimal features were remained. The radiomics combined ML models were yielded similar mean areas under the receiver operating characteristic curve of 0.808 (95% CI 0.754 to 0.861) for RF, 0.802 (95% CI 0.748 to 0.856) for radial basis kernel function-based SVM, 0.792 (95% CI 0.737 to 0.847) for MLP, 0.792 (95% CI 0.736 to 0.848) for Linear-SVM and 0.787 (95% CI 0.730 to 0.844) for LR, respectively. Combining radiomics with ML models can be an efficient, noninvasive, economical, and reliable technique for evaluating invisible microscopic AIS on NCCT and assisting radiologists to make clinical decisions.
Collapse
Affiliation(s)
- Kui Sun
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Rongchao Shi
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Xinxin Yu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing Wu Road, No. 324, Jinan, 250021, Shandong, China
| | - Ying Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing Wu Road, No. 324, Jinan, 250021, Shandong, China
| | - Wei Zhang
- Department of Radiology, Wangjing Hospital of CACMS, Beijing, 100102, China
| | - Xiaoxia Yang
- Department of Radiology, The Third People's Hospital of Datong, Datong, 037000, Shanxi, China
| | - Mei Zhang
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, Shandong, China
| | - Jian Wang
- Department of Radiology, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, Shandong, China
| | - Shu Jiang
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, 250014, Shandong Province, China
| | - Haiou Li
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Bing Kang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing Wu Road, No. 324, Jinan, 250021, Shandong, China
| | - Tong Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing Wu Road, No. 324, Jinan, 250021, Shandong, China
| | - Shuying Zhao
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China
| | - Yu Ai
- Department of Otolaryngology-Head and Neck Surgery, Cheeloo College of Medicine, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
| | - Jianfeng Qiu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Haiyan Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing Wu Road, No. 324, Jinan, 250021, Shandong, China.
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing Wu Road, No. 324, Jinan, 250021, Shandong, China.
| |
Collapse
|
49
|
He X, Yuan X, Shu Q, Gao Y, Chen Y, Liu Y, Xu J, Zhang Y, Cao G. Therapeutic effects of traditional Chinese medicine Hua-Feng-Dan in a rat model of ischemic stroke involve renormalization of gut microbiota. Front Pharmacol 2025; 16:1485340. [PMID: 39931688 PMCID: PMC11808003 DOI: 10.3389/fphar.2025.1485340] [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: 08/23/2024] [Accepted: 01/02/2025] [Indexed: 02/13/2025] Open
Abstract
Hua-Feng-Dan is a traditional Chinese medicine used to treat ischemic stroke, but little is known about its therapeutic mechanism. This study explored whether and how the mechanism involves readjustment of gut microbiota. Rats were subjected to middle cerebral artery occlusion as a model of ischemic stroke or to sham surgery, then treated or not with Hua-Feng-Dan. The different groups of animals were compared in terms of neurological score, cerebral infarct volume, brain edema, brain and gut histopathology to assess stroke severity. They were also compared in terms of indices of intestinal barrier permeability, inflammation and oxidative stress, brain metabolites as well as composition of the gut microbiota and their metabolites. Hua-Feng-Dan significantly reduced cerebral infarct volume and brain water content and improved neurological score, ischemic brain histopathology, and gut histopathology. It partially reversed stroke-induced intestinal barrier disruption and leakage, inflammation, dyslipidemia and oxidative stress, as well as the stroke-induced increase in pathogenic gut microbiota (e.g., Escherichia-Shigella, Enterococcus, Clostridium_innocuum_group) and decrease in beneficial microbiota (e.g., Lachnospiraceae, unclassified__f__Lachnospiracea and Ruminococcus_torques_group). The treatment altered levels of 39 and 38 metabolites produced during gut microbial and brain tissue metabolism respectively, mainly of amino acids, nucleosides, short-chain fatty acids, and essential fatty acids. Levels of factors related to inflammation and intestinal barrier permeability correlated positively with relative abundance of Escherichia-Shigella and Clostridium_innocuum_group, and negatively with 4-(glutamylamino) butanoate, 2-hydroxy-3-methylbutyric acid, dihomo-α-linolenic acid, dihomolinoleic acid, and 10-nitrolinoleic acid. Conversely, levels of 4-(glutamylamino) butanoate, 2-hydroxy-3-methylbutyric acid, and 10-nitrolinoleic acid correlated positively with relative abundance of unclassified__f__Lachnospiracea. Our results suggest that Hua-Feng-Dan may mitigate ischemic stroke injury by renormalizing gut microbiota and restoring gut barrier function, gut metabolism, thereby helping to alleviate inflammatory, neurological damage, and brain metabolic disorders.
Collapse
Affiliation(s)
- Xiaoxia He
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaofeng Yuan
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qilin Shu
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yayang Gao
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Youli Chen
- Zunyi Liao Yuan He Tang Pharmaceutical, Zunyi, China
| | - Yao Liu
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- National Engineering Technology Research Center for Miao Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jian Xu
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- National Engineering Technology Research Center for Miao Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yongping Zhang
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- National Engineering Technology Research Center for Miao Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Guoqiong Cao
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- National Engineering Technology Research Center for Miao Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
50
|
Zhu X, Zhang Z, Zhu Y, Chen Y, Li W, Xu H, Chen X. Comprehensive analysis of autophagy status and its relationship with immunity and inflammation in ischemic stroke through integrated transcriptomic and single-cell sequencing. Genes Immun 2025:10.1038/s41435-025-00320-y. [PMID: 39827328 DOI: 10.1038/s41435-025-00320-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Ischemic stroke (IS) significantly impacts patients' health and quality of life, with the roles of autophagy and autophagy-related genes in IS still not fully understood. In this study, IS datasets were retrieved from the GEO database. Autophagy-related genes(ARGs) were identified and screened for differential expression. A prediction model was constructed using machine learning algorithm. WGCNA was employed to analyze differential regulation modules among different clusters of stroke patients. The analysis results were validated using single-cell sequencing data. Finally, autophagy hub genes were validated in an external cohort and an IS mouse model. We observed suppressed autophagy states in IS patients. A diagnostic model with good clinical efficacy for stroke diagnosis was constructed based on the selected key genes (AUC = 0.87). Consensus clustering identified two IS subtypes with distinct gene expression patterns and immune cell infiltration. scRNA-seq data analysis confirmed downregulation of pexophagy in IS. CellChat analysis identified key signaling pathways and intercellular interactions related to pexophagy. Validation in an external cohort and IS mouse model confirmed differential gene expression, supporting the involvement of pexophagy in IS pathogenesis. The identified key genes, molecular subtypes, and cellular interactions provide a foundation for further research into targeted therapies and precision medicine approaches for IS patients.
Collapse
Affiliation(s)
- Xiaole Zhu
- Department of Emergency, Jiangsu Province Hospital and The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongman Zhang
- Department of Emergency, Jiangsu Province Hospital and The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Zhu
- Department of Emergency, Jiangsu Province Hospital and The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanlong Chen
- Department of Emergency, Jiangsu Province Hospital and The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Li
- Department of Emergency, Jiangsu Province Hospital and The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huae Xu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xufeng Chen
- Department of Emergency, Jiangsu Province Hospital and The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|