1
|
Zhou Q, Huang X, Chen Z, Wang F, Xie L, Sun Q, Du J, Lin J, Li B, Li L. Mechanism of Rhizoma Polygonati in the treatment of Alzheimer's disease based on network pharmacology and molecular docking. Brain Res 2025; 1862:149680. [PMID: 40412586 DOI: 10.1016/j.brainres.2025.149680] [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: 11/27/2024] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVE The therapeutic mechanisms of Rhizoma Polygonati (RP) on Alzheimer's disease (AD) were explored using network pharmacology methods and in vitro experiments for validation. MATERIALS AND METHODS First, the main active ingredients and target proteins of RP were screened using Traditional Chinese Medicine Systems Pharmacology (TCMSP) and UniProt protein database. AD-related targets were predicted using the DisGeNET database. Subsequently, Protein-protein interaction (PPI) networks and core targets were analyzed using STRING. DAVID was utilized for GO annotation, while KEGG plug-in was employed to perform enrichment analysis of KEGG pathways. AutoDockTools were examined molecular docking. And the RP mechanism on AD was confirmed in vitro experimentation. RESULTS Screening identified 8 active ingredients, 76 potential targets, and 3397 CE-related genes, with 58 overlapping targets. 4 target proteins were analyzed through the PPI networks. The RP and AD shared 451 GO biological process items and 150 KEGG signal pathways. Molecular docking results showed that diosgenin (Dio) had strong binding abilities to AKT1 and Caspase 3. Dio inhibited apoptosis through AKT1/Caspase 3 pathway in the glutamate-induced SH-SY5Y cells in vitro. CONCLUSION The study revealed RP's potential mechanisms in treating AD, offering a theoretical basis for clinical use, by integrating network pharmacology with in vitro experiments.
Collapse
Affiliation(s)
- Qiong Zhou
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | | | - Zihao Chen
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Fuwei Wang
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Lihua Xie
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Qiang Sun
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jikun Du
- Central Research Laboratory, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen 518104, China.
| | - Jiantao Lin
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| | - Baohong Li
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| | - Li Li
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
2
|
Wang H, Chao L, Shen S, You P, Li L, Chen X, Hong Z, Chai Y. Exploring the pharmacological mechanism of Bu-Wang San on Alzheimer's disease through multiple GEO datasets of the human hippocampus, network pharmacology, and metabolomics based on GC-MS and UPLC-Q/TOF-MS. JOURNAL OF ETHNOPHARMACOLOGY 2025; 350:119994. [PMID: 40389089 DOI: 10.1016/j.jep.2025.119994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 05/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bu-Wang San (BWS) is a prominent traditional Chinese medicine known for calming the mind and promoting intelligence. It has been reported to improve learning and memory, enhance memory ability, and promote synaptic plasticity. However, the complexity of the material basis and the diversity of therapeutic targets of BWS on Alzheimer's disease (AD) have not been elucidated. AIM OF THE STUDY This study aimed to investigate the therapeutic material basis and the mechanism of BWS in AD treatment by comprehensively analyzing multiple GEO datasets of the human hippocampus, network pharmacology, and multi-platform metabolomics validation. MATERIALS AND METHODS Three GEO datasets of the human hippocampus were utilized to identify AD-associated targets using weighted gene co-expression network analysis (WGCNA) and differential analysis. Network pharmacology analyses were performed to investigate BWS's therapeutic material basis and predict the therapeutic targets of BWS on AD. A rat model was induced through the concurrent administration of AlCl3 and D-galactose to validate BWS's therapeutic potential and underlying mechanisms in AD. To validate the results of GEO data mining and network pharmacology, a comprehensive metabolomics approach integrating gas chromatography-mass spectrometry (GC-MS) and ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF/MS) was conducted on rat serum samples to uncover potential metabolic alterations and their associated pathways. RESULTS A total of 6367 genes were selected as AD drug targets through WGCNA analysis and enrichment analysis of disease-associated gene expression profiles in the GEO database. Network pharmacology was performed in this study for the identification of potential interactions between the components of BWS and its targets, TP53, STAT3, EGFR, MAOA, NOS3, PPARG, PRKCA, MAPK8, AChE, ARG1, among others, which were among the top 25 highest probable targets of BWS acting on AD. The multi-platform metabolomics indicated that amino sugar and nucleotide sugar metabolism, glycine, serine and threonine metabolism pathways, and other pathways may be associated with the AD model based on AlCl3 and D-galactose. The comparison of differential metabolites between the AD model group and the BWS intervention group revealed that 66 of the 97 differential metabolites exhibited a pullback trend, indicating a potential therapeutic effect of BWS on these metabolites. CONCLUSION This study builds a systematic strategy combining GEO datasets, network pharmacology, and multi-platform metabolomics and provides valuable insights into the pharmacological mechanism of BWS on AD. The results suggest that BWS may exert its therapeutic effects on AD by modulating the amino sugar and nucleotide sugar metabolism, glycerophospholipid metabolism, glycine, serine and threonine metabolism pathway and acting on the drug targets of ARG1, MAOA, AChE, XDH, GAD2 et al. This strategy provides a deep understanding of the molecular mechanisms of herbal medicine in treating AD at a systematic level.
Collapse
Affiliation(s)
- Hui Wang
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Liang Chao
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Shuqi Shen
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Piaoxue You
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | - Ling Li
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Xiaofei Chen
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
| | - Zhanying Hong
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
| | - Yifeng Chai
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
3
|
Han J, Zhang X, Kang L, Guan J. Extracellular vesicles as therapeutic modulators of neuroinflammation in Alzheimer's disease: a focus on signaling mechanisms. J Neuroinflammation 2025; 22:120. [PMID: 40281600 PMCID: PMC12023694 DOI: 10.1186/s12974-025-03443-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-beta (Aβ) plaques and tau tangles, which contribute significantly to neuroinflammation, a central driver of disease pathogenesis. The activation of microglia and astrocytes, coupled with the complex interactions between Aβ and tau pathologies and the innate immune response, leads to a cascade of inflammatory events. This process triggers the release of pro-inflammatory cytokines and chemokines, exacerbating neuronal damage and fostering a cycle of chronic inflammation that accelerates neurodegeneration. Key signaling pathways, such as nuclear factor-kappa B (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), mitogen-activated protein kinase (MAPK), and phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), are involved in regulating the production of these inflammatory mediators, offering potential therapeutic targets for AD. Recently, extracellular vesicles (EVs) have emerged as a promising tool for AD therapy, due to their ability to cross the blood-brain barrier (BBB) and deliver therapeutic agents. Despite challenges in standardizing EV-based therapies and ensuring their safety, EVs offer a novel approach to modulating neuroinflammation and promoting neuroregeneration. This review aims to highlight the intricate relationship between neuroinflammation, signaling pathways, and the emerging role of EV-based therapeutics in advancing AD treatment strategies.
Collapse
Affiliation(s)
- Jingnan Han
- Department of Ophthalmology, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110000, China
| | - Xue Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, 110000, China
| | - Longdan Kang
- Department of Ophthalmology, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110000, China.
| | - Jian Guan
- Department of Ophthalmology, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110000, China.
| |
Collapse
|
4
|
Wang M, Wan M, Liu M, Zhou W, Zhang X, Liu W, Liu Y, Jiang S, Shang E, Duan J. Integrated network pharmacology and metabolomics analysis to reveal the potential mechanism of Ershen Wan in ameliorating ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119690. [PMID: 40158827 DOI: 10.1016/j.jep.2025.119690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ershen Wan (ESW), a classic traditional Chinese medicine (TCM) prescription composed of Psoralea corylifolia Linn. and Myristica fragrans Houtt., has been applied to treat gastrointestinal disorders in clinical practices for thousands of years. However, its potential molecular mechanism in alleviating ulcerative colitis (UC) remains to be elusive. AIM OF THE STUDY The purpose of the study is to explore the underlying mechanism of ESW in treating UC. MATERIALS AND METHODS The protective effect of ESW on dextran sodium sulfate (DSS)-induced UC mice was assessed by body weight, disease activity index (DAI), colon length, colon tissue pathology, and colonic inflammatory factors. Furthermore, network pharmacology was applied to dissect the possible targets and biological pathways regulated by ESW. The plasma and fecal metabolomics were comprehensively analyzed by UPLC-Q-TOF/MS. Subsequently, an efficient and feasible approach integrating network pharmacology, metabolomics, and molecular docking was used to explore the key targets obtained from the metabolite-reaction-enzyme-gene network. And the effect of ESW on the MAPK signaling mediated intestinal epithelial cell apoptosis was further investigated by in vitro and in vivo experiments. RESULTS ESW could notably alleviate colon injury and inflammation of UC mice. Network pharmacology suggested that the bioactive components of ESW could mainly modulate signaling pathways associated with inflammation and metabolism. Consistently, plasma and fecal metabolomics further indicated that ESW could regulate the metabolic pathways of arachidonic acid, linoleic acid, sphingolipid, tryptophan, and glycerophospholipid. And the combined analysis of network pharmacology and metabolomics revealed that 14 pivotal targets were modulated by ESW, including PTGS1, PTGS2, CYP1A1, FADS1, CBR1, ALOX5, EPHX1, EPHX2, HPGD, PLA2G1B, PLA2G7, MGLL, ACHE, and SPHK1. Additionally, molecular docking suggested that bioactive components of ESW could bind well to these potential targets. And in vitro and in vivo experiments further verified that ESW could markedly ameliorate pathological symptoms of UC mice through inhibiting MAPK signaling mediated colonic epithelial cell apoptosis. CONCLUSION Collectively, these findings indicated that ESW could effectively alleviate the pathological symptoms of UC mice, mainly involving in the modulation of lipid and amino acid metabolism pathways, and the suppression of MAPK signaling-mediated apoptosis. In this study, the potential mechanism of ESW for the treatment of UC was first clarified, which provided a solid scientific foundation for its clinical application. Notably, the proposed strategy facilitated a comprehensive prediction and validation of the efficacy and molecular mechanism of TCMs, and also provided a novel approach for revealing the intricate biological pathogenesis of diseases.
Collapse
Affiliation(s)
- Mingyang Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Meiyu Wan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Meijuan Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Wenwen Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Xiaoxiao Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Weijie Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Ying Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Erxin Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| |
Collapse
|
5
|
Shi X, Wang Z, Liu Z, Lin Q, Huang M, Lim TY, Li X, Wang T. Qingqi Guxue Decoction induces S cell cycle arrest to inhibit replication of severe fever with thrombocytopenia syndrome virus. Virol Sin 2025; 40:260-274. [PMID: 40157606 DOI: 10.1016/j.virs.2025.03.011] [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: 09/25/2024] [Accepted: 03/25/2025] [Indexed: 04/01/2025] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is a novel emerging acute infectious disease caused by severe fever with thrombocytopenia syndrome virus (SFTSV), characterized by high fever and thrombocytopenia. It has been proved that traditional Chinese medicine (TCM) has displayed definite therapeutic effects on viral hemorrhagic fever, indicating its potential to treat SFTS. In this study, SFTS-relative key targets were predicted via gene ontology (GO) analysis and kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis. Molecular docking was then used to select stable binders. Molecules matched TCMs were identified, and a new prescription, Qingqi Guxue decoction (QQGX), was formulated to clear heat and nourish blood, with a resulting drug composition network. We explored the optimal drug proportion for QQGX. Through an in-depth study of molecular mechanisms, we found that QQGX induces S phase arrest by promoting the degradation of cyclin A2 (CCNA2) and cyclin-dependent kinase 2 (CDK2), thereby inhibiting SFTSV replication. Finally, we verified the effectiveness and safety of QQGX based on the mouse liver bile duct organoid model infected with SFTSV. In summary, our study prepared a TCM decoction using the method of network pharmacology. This decoction has a significant inhibitory effect on the replication of SFTSV and provides a new treatment strategy for hemorrhagic fever with TCM.
Collapse
Affiliation(s)
- Xixi Shi
- School of Life Sciences, Tianjin University, Tianjin 300110, China
| | - Zining Wang
- School of Life Sciences, Tianjin University, Tianjin 300110, China
| | - Zixiang Liu
- School of Life Sciences, Tianjin University, Tianjin 300110, China
| | - Qinting Lin
- School of Life Sciences, Tianjin University, Tianjin 300110, China
| | - Mengqian Huang
- School of Life Sciences, Tianjin University, Tianjin 300110, China
| | - Tze Yean Lim
- School of Life Sciences, Tianjin University, Tianjin 300110, China
| | - Xiaoyan Li
- Tianjin Centers for Disease Control and Prevention, Tianjin 300022, China; Tianjin Key Laboratory of Pathogenic Microbiology of Infectious Disease, Tianjin 300011, China
| | - Tao Wang
- School of Life Sciences, Tianjin University, Tianjin 300110, China; Tianjin Key Laboratory of Pathogenic Microbiology of Infectious Disease, Tianjin 300011, China.
| |
Collapse
|
6
|
Qin G, Song R, Sun J, Dai J, Wang W, Meng F, Wang D, Liu Z, Sun B, Li C. Unveiling the Therapeutic Potential of Banxia Xiexin Decoction in Alzheimer's Disease: Insights From Network Pharmacology and Experimental Validation. Drug Des Devel Ther 2025; 19:2133-2155. [PMID: 40134954 PMCID: PMC11934878 DOI: 10.2147/dddt.s499852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Background Alzheimer's disease (AD) is associated with various pathological states for which there is no effective treatment. First documented in the Eastern Han Dynasty's medical classic, "Treatise on Febrile and Miscellaneous Diseases" (200-210 Anno Domini), Banxia Xiexin Decoction (BXD) stands as a quintessential approach to treating spleen ailments. Recent studies have shown BXD's effectiveness in mitigating memory impairment associated with AD. Yet, the precise mechanisms underlying BXD's action against AD require further exploration. Aim of the Study To explore the important components of BXD in exerting anti-AD effects and the underlying molecular mechanisms using network pharmacology, metabolomics analysis, and in vitro and in vivo validation strategies. Initially, candidates for BXD's application in AD therapy were identified through extensive database searches, followed by an analysis of protein-protein interactions (PPI). To elucidate BXD's therapeutic pathways in AD, we engaged in Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) assessments. Further, we delved into BXD's primary constituents through ultra-high-pressure liquid chromatography coupled with Q Exactive mass spectrometry and molecular docking techniques. Finally, AD-associated Aβ42-SY5Y cells and APPswe/PS1dE9 (APP/PS1) transgenic mice models were utilized to further determine the activity and mechanisms of BXD through various molecular or phenotypic assays and metabolomics analysis. Results Our findings identified the PI3K/Akt signaling pathways as central to BXD's effects. Using in vitro and in vivo models, we found the activity of BXD against AD to be mediated by the suppression of neuroinflammation and apoptosis, accompanied by activation of the PI3K/Akt pathway. Finally, we observed robust changes in metabolite levels in the plasma of BXD-treated APP/PS1 mice. Conclusion Through systematic data analysis and experimental validation, the therapeutic advantages and fundamental molecular mechanisms of BXD in treating AD were revealed. These findings underscore the promising prospects and compelling potential of BXD, which targets the PI3K/Akt signaling pathway and inflammation, apoptosis, as a therapeutic strategy for improving AD.
Collapse
Affiliation(s)
- Gaofeng Qin
- Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
- Postdoctoral Research Mobile Station, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Rongqiang Song
- Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Jingyi Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Juanjuan Dai
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Wentao Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Fantao Meng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Dan Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Zhe Liu
- Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Baoliang Sun
- Second AfFIliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, People’s Republic of China
| | - Chen Li
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| |
Collapse
|
7
|
Hou X, Liang X, Zhao X, Shi Y, Zhuo F, Tong X, Yang X, Zhai Q, Wang J, Guo Q, Tu P, Zeng K, Zhang Q. Uncaria rhynchophylla alkaloid extract exerts neuroprotective activity against Parkinson's disease via activating mitophagy with the involvement of UCHL1. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119009. [PMID: 39471877 DOI: 10.1016/j.jep.2024.119009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/25/2024] [Accepted: 10/26/2024] [Indexed: 11/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Uncaria rhynchophylla (Miq.) Miq. ex Havil. (UR), a traditional Chinese medicinal plant, plays an active role in neuroprotection. Clinical medication and modern pharmacological studies have proved the efficacy of UR against Parkinson's disease (PD), with alkaloids being recognized as the main bioactive components. But the therapeutic effect and mechanism of U. rhynchophylla alkaloid extract (URA) against PD need further exploration. AIM OF STUDY The study aimed to investigate the therapeutic effect and potential mechanism of URA on PD. MATERIALS AND METHODS LC-MS methodology was used to evaluate the chemical constituents of URA. The anti-PD activity of URA in vivo was measured on the mouse and rat models of PD. Neuroprotective effect of URA on PC12 cells was measured by MTT assay. Dopamine (DA) and its metabolites were detected by LC-MS for probing the protection ability on dopaminergic neurons. The differentially expressed proteins between model group and URA administrated group were analyzed by proteomics, suggesting oxidative phosphorylation as possible pathway of URA. Considering the critical role of mitochondria in oxidative phosphorylation, JC-1 staining, MitoSOX staining, transmission electron microscopy (TEM) observation and adenosine triphosphate (ATP) levels detection were used to analyze the effects of URA on mitochondrial morphology and function. Biolayer interferometry (BLI) was used to search for the possible UCHL1-bonding compounds in URA. RESULTS URA significantly mitigated the behavioural defects by improving coordination, shortening the time to climb down the whole pole (T-LA) and increasing the forelimbs' muscle strength of MPTP-induced PD mice and 6-OHDA-induced PD rats. In addition, URA improved tyrosine hydroxylase expression in dopaminergic neurons by immunohistochemistry (IHC) staining, and thus increased the neurotransmitter levels of DA and relevant metabolites. Furthermore, URA promoted mitophagy as reflected by a significant decrease in reactive oxygen species (ROS) generation, an increase in ATP levels and clearance of damaged mitochondria. Subsequently, Ubiquitin C-terminal hydrolase 1 (UCHL1), which is associated with the mitochondrial dysfunction in PD, is suggested to be a promising target based on the proteomics result, and proved by the blocked protective effects of URA by UCHL1 inhibitor. Furthermore, hirsuteine (HTE) was identified as a potential active compound of URA binding to UCHL1 by BLI, and the binding capacity and site were verified by surface plasmon resonance (SPR) and molecular docking. CONCLUSION This work demonstrates that URA exerts effective neuroprotective activity against PD via activation of mitophagy with the involvement of UCHL1, and HTE may be a potential active compound of URA.
Collapse
Affiliation(s)
- Xingzi Hou
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Xiaomin Liang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Xin Zhao
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, PR China.
| | - Fangfang Zhuo
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Xinnuo Tong
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Xunfang Yang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Qi Zhai
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Jie Wang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Qiang Guo
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, PR China.
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Qingying Zhang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| |
Collapse
|
8
|
Xiao Y, Zhang S, Zhuo H, Zhang X, Zhu K, Chen W, You G, Chen H, Luo Q, Zhou H, Chen G. Dietary Natural Melanin Nanozymes Delay Aging and Ameliorate Neurodegeneration via Improving Gut Microbiota and Redox Homeostasis. ACS OMEGA 2025; 10:3610-3621. [PMID: 39926539 PMCID: PMC11800042 DOI: 10.1021/acsomega.4c08419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 02/11/2025]
Abstract
Aging is an inevitable multifactor process that causes a decline in organ function and increases the risk of age-related diseases and death. Thus, the development of highly effective and safe therapeutic strategies to delay aging and age-related diseases is urgently required. In this study, we isolated natural melanin nanozymes (NMNs) from the ink sacs of live octopuses. The NMNs exhibited excellent superoxide-dismutase-mimicking and radical scavenging activities. In SAMP8 mice, treatment with NMNs improved their cognition and memory functions while restoring their aging-impaired liver function and lipid metabolism, thereby prolonging their lifespan. Moreover, the NMNs reversed metabolic changes in their aged brains and reconstructed their gut microbiota composition by enhancing microbial community diversity. Our findings indicate that NMNs treatment could be a promising approach for delaying aging and preventing age-associated physiological decline in humans.
Collapse
Affiliation(s)
- Yao Xiao
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Shikun Zhang
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Hailong Zhuo
- Department
of Transfusion, The Fifth Medical Center
of Chinese PLA General Hospital, Beijing 100071, China
| | - Xiaoyong Zhang
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Kai Zhu
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Wanyi Chen
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Guoxing You
- Academy
of Military Medical Sciences, Beijing 100850, China
| | | | - Qun Luo
- Department
of Transfusion, The Fifth Medical Center
of Chinese PLA General Hospital, Beijing 100071, China
| | - Hong Zhou
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Gan Chen
- Academy
of Military Medical Sciences, Beijing 100850, China
| |
Collapse
|
9
|
Liu Z, Jia J. Omaveloxolone Ameliorates Cognitive Deficits by Inhibiting Apoptosis and Neuroinflammation in APP/PS1 Mice. Mol Neurobiol 2025; 62:2191-2202. [PMID: 39088030 DOI: 10.1007/s12035-024-04361-8] [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: 02/21/2024] [Accepted: 07/09/2024] [Indexed: 08/02/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease associated with aging, characterized by progressive cognitive impairment and memory loss. However, treatments that delay AD progression or improve its symptoms remain limited. The aim of the present study was to investigate the therapeutic effects of omaveloxolone (Omav) on AD and to explore the underlying mechanisms. Thirty-week-old APP/PS1 mice were selected as an experimental model of AD. The spatial learning and memory abilities were tested using the Morris water maze. Amyloid-beta (Aβ) deposition in the brains was measured using immunohistochemistry. Network pharmacological analyses and molecular docking were conducted to gain insights into the therapeutic mechanisms of Omav. Finally, validation analyses were conducted to detect changes in the associated pathways and proteins. Our finding revealed that Omav markedly rescued cognitive dysfunction and reduced Aβ deposition in the brains of APP/PS1 mice. Network pharmacological analysis identified 112 intersecting genes, with CASP3 and MTOR emerging as the key targets. In vivo validation experiments indicated that Omav attenuated neuronal apoptosis by regulating apoptotic proteins, including caspase 3, Bax, and Bcl-2. Moreover, Omav suppressed neuroinflammation and induced autophagy by inhibiting the phosphorylation of mTOR. These findings highlight the therapeutic efficacy of Omav in AD and that its neuroprotective effects were associated with inhibiting neuronal apoptosis and regulating neuroinflammation.
Collapse
Affiliation(s)
- Zhaojun Liu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Changchun Street 45, Beijing, PR China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Changchun Street 45, Beijing, PR China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, PR China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, PR China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, PR China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, PR China.
| |
Collapse
|
10
|
Yan X, Inta A, Yang X, Pandith H, Disayathanoowat T, Yang L. An Investigation of the Effect of the Traditional Naxi Herbal Formula Against Liver Cancer Through Network Pharmacology, Molecular Docking, and In Vitro Experiments. Pharmaceuticals (Basel) 2024; 17:1429. [PMID: 39598341 PMCID: PMC11597843 DOI: 10.3390/ph17111429] [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: 10/02/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: The formula Chong-Lou-Yao-Fang (CLYF) is an herbal medicinal formulation developed by the indigenous Naxi people for treating liver cancer. This study was to reveal the biological activity, potential targets, and molecular mechanisms of CLYF for cancer treatment. Methods: Network pharmacology, microarray data analysis, survival analysis, and molecular docking were employed to predict potential compounds, targets, and pathways for the treatment of liver cancer. In vitro experiments and Western blot validation were conducted to confirm these predictions. Results: 35 key compounds and 20 core targets were screened from CLYF, involving signaling pathways for PI3K-Akt, MAPK, hepatitis B and C, which were effective for liver cancer treatment. Microarray data analysis and survival analysis indicated that EGFR and TP53 serve as promising biomarkers for diagnosis and prognosis in liver cancer. Molecular docking revealed stable binding between EGFR, TP53, and AKT1 with active ingredients. Cell experiments confirmed that CLYF-A suppressed cell proliferation, induced apoptosis, and caused cell cycle arrest in HepG2 cells, which were associated with a loss of mitochondrial membrane potential. Compared to the control group, the relative protein expression levels of EGFR and AKT1 significantly decreased following treatment with CLYF-A, while TP53 levels increased significantly. Conclusions: Verification of the anticancer activity of CLYF and its potential mechanisms may have important implications for anticancer therapies. Our results may provide a scientific basis for the clinical use of CLYF for cancer treatment and have important implications for developing pharmaceutical preparations, which also need more pharmacological experiments, clinical experiments, and in vivo experiments.
Collapse
Affiliation(s)
- Xiuxiang Yan
- Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; (X.Y.); (X.Y.)
- Department of Biology, Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand; (A.I.); (H.P.)
| | - Angkhana Inta
- Department of Biology, Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand; (A.I.); (H.P.)
| | - Xuefei Yang
- Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; (X.Y.); (X.Y.)
- Yunnan International Joint Laboratory of Southeast Asia Biodiversity Conservation, Menglun 666303, China
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Yezin, Nay Pyi Taw 05282, Myanmar
| | - Hataichanok Pandith
- Department of Biology, Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand; (A.I.); (H.P.)
| | - Terd Disayathanoowat
- Department of Biology, Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai 50200, Thailand; (A.I.); (H.P.)
| | - Lixin Yang
- Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; (X.Y.); (X.Y.)
- Yunnan International Joint Laboratory of Southeast Asia Biodiversity Conservation, Menglun 666303, China
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Yezin, Nay Pyi Taw 05282, Myanmar
| |
Collapse
|
11
|
Gao Y, Zhao P, Wang C, Fang K, Pan Y, Zhang Y, Miao Z, Wang M, Wei M, Zou W, Liu M, Peng K. Buqi Huoxue Tongnao prescription protects against chronic cerebral hypoperfusion via regulating PI3K/AKT and LXRα/CYP7A1 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155844. [PMID: 38959552 DOI: 10.1016/j.phymed.2024.155844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/01/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) has been confirmed as one of the pathogenesis underlying vascular cognitive impairment. A series of pathological changes, including inflammation, oxidative stress, and apoptosis, are involved in this pathophysiology and contribute to cognitive impairment and neuropathological alterations. The traditional Chinese medicine (TCM) of Buqi Huoxue Tongnao (BQHXTN) prescription possesses a remarkable clinical efficacy for treating patients with CCH, but still lacks a scientific foundation for its pharmacological mechanisms. PURPOSE To investigate the role and underlying mechanism of the effects of BQHXTN on CCH both in vitro and in vivo. METHODS In this study, we established a two-vessel occlusion (2-VO) induced CCH model in Sprague-Dawley rats, an oxygen-glucose deprivation model in BV2 cells, and a steatosis cell model in L02 cells to reveal the underlying mechanisms of BQHXTN by behavioral test, histopathological analysis and the detection of pro-inflammatory cytokine, apoptotic factors and reactive oxide species. Donepezil hydrochloride and Buyang Huanwu decoction were used as positive drugs. RESULTS Compared with the 2-VO group, BQHXTN treatment at three doses significantly enhanced the memory and learning abilities in the Y-maze and novel object recognition tests. The hematoxylin-eosin staining indicated that BQHXTN protected against hippocampal injury induced by CCH. Of note, in both in vivo and in vitro experiments, BQHXTN prominently inhibited the production of IL-1β, TNF-α, cleaved-caspase 3, and iNOS by regulating the PI3K/AKT pathway, consequently exerting anti-inflammatory, anti-apoptotic, and antioxidant effects. Moreover, it provided the first initial evidence that BQHXTN treatment mitigated dyslipidemia by increasing the LXRα/CYP7A1 expression, thereby delaying the neuropathological process. CONCLUSION In summary, these findings firstly revealed the pharmacodynamics and mechanism of BQHXTN, that is, BQHXTN could alleviate cognitive impairment, neuropathological alterations and dyslipidemia in CCH rats by activating PI3K/AKT and LXRα/CYP7A1 signaling pathways, as well as providing a TCM treatment strategy for CCH.
Collapse
Affiliation(s)
- Yinhuang Gao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China; Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peng Zhao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China
| | - Chunyan Wang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China
| | - Keren Fang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China
| | - Yueqing Pan
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Zhang
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhishuo Miao
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Meirong Wang
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Minlong Wei
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wei Zou
- Changsha Research and Development Center on Obstetric and Gynecologic Traditional Chinese Medicine Preparation, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
| | - Menghua Liu
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China; Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Kang Peng
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China.
| |
Collapse
|
12
|
Zhang D, Qin H, Chen W, Xiang J, Jiang M, Zhang L, Zhou K, Hu Y. Utilizing network pharmacology, molecular docking, and animal models to explore the therapeutic potential of the WenYang FuYuan recipe for cerebral ischemia-reperfusion injury through AGE-RAGE and NF-κB/p38MAPK signaling pathway modulation. Exp Gerontol 2024; 191:112448. [PMID: 38697555 DOI: 10.1016/j.exger.2024.112448] [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: 02/21/2024] [Revised: 04/13/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Stroke is a debilitating condition with high morbidity, disability, and mortality that significantly affects the quality of life of patients. In China, the WenYang FuYuan recipe is widely used to treat ischemic stroke. However, the underlying mechanism remains unknown, so exploring the potential mechanism of action of this formula is of great practical significance for stroke treatment. OBJECTIVE This study employed network pharmacology, molecular docking, and in vivo experiments to clarify the active ingredients, potential targets, and molecular mechanisms of the WenYang FuYuan recipe in cerebral ischemia-reperfusion injury, with a view to providing a solid scientific foundation for the subsequent study of this recipe. MATERIALS AND METHODS Active ingredients of the WenYang FuYuan recipe were screened using the traditional Chinese medicine systems pharmacology database and analysis platform. Network pharmacology approaches were used to explore the potential targets and mechanisms of action of the WenYang FuYuan recipe for the treatment of cerebral ischemia-reperfusion injury. The Middle Cerebral Artery Occlusion/Reperfusion 2 h Sprague Dawley rat model was prepared, and TTC staining and modified neurological severity score were applied to examine the neurological deficits in rats. HE staining and Nissl staining were applied to examine the pathological changes in rats. Immunofluorescence labeling and Elisa assay were applied to examine the expression levels of certain proteins and associated factors, while qRT-PCR and Western blotting were applied to examine the expression levels of linked proteins and mRNAs in disease-related signaling pathways. RESULTS We identified 62 key active ingredients in the WenYang FuYuan recipe, with 222 highly significant I/R targets, forming 138 pairs of medication components and component-targets, with the top five being Quercetin, Kaempferol, Luteolin, β-sitosterol, and Stigmasterol. The key targets included TP53, RELA, TNF, STAT1, and MAPK14 (p38MAPK). Targets related to cerebral ischemia-reperfusion injury were enriched in chemical responses, enzyme binding, endomembrane system, while enriched pathways included lipid and atherosclerosis, fluid shear stress and atherosclerosis, AGE-RAGE signaling in diabetic complications. In addition, the main five active ingredients and targets in the WenYang FuYuan recipe showed high binding affinity (e.g. Stigmasterol and MAPK14, total energy <-10.5 Kcal/mol). In animal experiments, the WenYang FuYuan recipe reduced brain tissue damage, increased the number of surviving neurons, and down-regulated S100β and RAGE protein expression. Moreover, the relative expression levels of key targets such as TP53, RELA and p38MAPK mRNA were significantly down-regulated in the WenYang FuYuan recipe group, and serum IL-6 and TNF-a factor levels were reduced. After WenYang FuYuan recipe treatment, the AGE-RAGE signaling pathway and downstream NF-kB/p38MAPK signaling pathway-related proteins were significantly modulated. CONCLUSION This study utilized network pharmacology, molecular docking, and animal experiments to identify the potential mechanism of the WenYang FuYuan recipe, which may be associated with the regulation of the AGE-RAGE signaling pathway and the inhibition of target proteins and mRNAs in the downstream NF-kB/p38MAPK pathway.
Collapse
Affiliation(s)
- Ding Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Hongling Qin
- Guangxi University of Chinese Medicine First Affiliated Hospital, Nanning, China
| | - Wei Chen
- Guangxi University of Chinese Medicine First Affiliated Hospital, Nanning, China
| | - Junjun Xiang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Minghe Jiang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Ling Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Keqing Zhou
- Guangxi University of Chinese Medicine, Nanning, China
| | - Yueqiang Hu
- Guangxi University of Chinese Medicine First Affiliated Hospital, Nanning, China.
| |
Collapse
|
13
|
Mi Y, Liang Y, Liu Y, Bai Z, Li N, Tan S, Hou Y. Integrated network pharmacology and experimental validation-based approach to reveal the underlying mechanisms and key material basis of Jinhua Qinggan granules against acute lung injury. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117920. [PMID: 38373663 DOI: 10.1016/j.jep.2024.117920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jinhua Qinggan granules (JHQG), the traditional Chinese formula come into the market in 2016, has been proved clinically effective against coronavirus disease. Acute lung injury (ALI) is a major complication of respiratory infection such as coronavirus and influenza virus, with a high clinical fatality rate. Macrophage activation-induced inflammatory response plays a crucial role in the pathogenesis of ALI. However, the participation of inflammatory response in the efficacy of JHQG and its material basis against ALI is still unknown. AIM OF THE STUDY The research aims to investigate the inflammatory response-involved efficacy of JHQG on ALI, explore the "ingredient-target-pathway" mechanisms, and searching for key material basis of JHQG by integrated network pharmacology and experimental validation-based approach. MATERIALS AND METHODS Lipopolysaccharide (LPS)-induced ALI mice was established to assess the protective impact of JHQG. Network pharmacology was utilized to identify potential targets of JHQG and investigate its action mechanisms related to inflammatory response in treating ALI. The therapeutic effect and mechanism of the primary active ingredient in JHQG was verified through high performance liquid chromatography (HPLC) and a combination of wet experiments. RESULTS JHQG remarkably alleviated lung damage in mice model via suppressing macrophage activation, and inhibiting pro-inflammatory mediator level, p-ERK and p-STAT3 expression, TLR4/NF-κB activation. Network pharmacology combined with HPLC found luteolin is the main effective component of JHQG, and it could interact with TLR4/MD2 complex, further exerting the anti-inflammatory property and the protective role against ALI. CONCLUSIONS In summary, our finding clarified the underlying mechanisms and material basis of JHQG therapy for ALI by integrated network pharmacology and experimental validation-based strategy.
Collapse
Affiliation(s)
- Yan Mi
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yusheng Liang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yeshu Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Zisong Bai
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China; School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning Province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning Province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China
| | - Shaowen Tan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China.
| |
Collapse
|
14
|
Zhang L, Zhang H, Niu X, Zhang X, Chen X, Lei S, Ma S, Sun Z. Liangxue-Qushi-Zhiyang Decoction Ameliorates DNCB-Induced Atopic Dermatitis in Mice through the MAPK Signaling Pathway Based on Network Pharmacology. ACS OMEGA 2024; 9:17931-17944. [PMID: 38680355 PMCID: PMC11044150 DOI: 10.1021/acsomega.3c09218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024]
Abstract
The traditional prescription of Liangxue-Qushi-Zhiyang decoction (LQZ) has been demonstrated to be efficacious in treating atopic dermatitis (AD), a chronic inflammatory skin disorder marked by intense itching, redness, rashes, and skin thickening. Nevertheless, there has been an inadequate systematic exploration of the potential targets, biological processes, and pathways for AD treatment through LQZ. The study objective was to evaluate the efficacy and possible mechanism of LQZ in AD mice. In our study, we identified the primary compounds of LQZ, analyzed hub targets, and constructed a network. Subsequently, the predicted mechanisms of LQZ in AD were experimentally studied and validated in vivo, as determined by network pharmacological analysis. A total of 80 serum components of LQZ were identified through ultra-performance liquid chromatography-electrospray ionization-tandem mass spectrometry (UPLC-ESI-MS/MS), among which 49 compounds were absorbed into the bloodstream. Our results indicated that LQZ targets six putative key factors in the MAPK signaling pathway, which play essential roles in AD, namely, EGFR, p-MAPK1/3, p-MAPK14, IL-1β, IL-6, and TNF-α. We observed spleen coefficient, dermatitis scores, and ear thickness were all downregulated in 2,4-dinitrochlorobenzene (DNCB)-induced mice after LQZ treatment. Histological analysis of the dorsal and ear skin further revealed that LQZ significantly decreased skin inflammation, epidermal thickness, and mast cell numbers compared to the DNCB group. Our study demonstrated the effectiveness of LQZ in reducing epidermal and dermal damage in a mouse model of AD. Furthermore, our findings suggest that downregulating the MAPK signaling pathway could be a potential therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Lili Zhang
- Beijing
University of Chinese Medicine Affiliated Third Hospital, Beijing 100029, China
| | - Huili Zhang
- Beijing
University of Chinese Medicine Dongfang Hospital, Beijing 100078, China
| | - Xiaoyu Niu
- Beijing
University of Chinese Medicine Affiliated Third Hospital, Beijing 100029, China
| | - Xuan Zhang
- Beijing
University of Chinese Medicine Affiliated Third Hospital, Beijing 100029, China
| | - Xingtong Chen
- Beijing
University of Chinese Medicine Affiliated Third Hospital, Beijing 100029, China
| | - Shengyi Lei
- Beijing
University of Chinese Medicine Affiliated Third Hospital, Beijing 100029, China
| | - Shengnan Ma
- Beijing
University of Chinese Medicine Affiliated Third Hospital, Beijing 100029, China
| | - Zhanxue Sun
- Beijing
University of Chinese Medicine Affiliated Third Hospital, Beijing 100029, China
| |
Collapse
|
15
|
Zou D, Huang X, Lan Y, Pan M, Xie J, Huang Q, Zeng J, Zou C, Pei Z, Zou C, Mao Y, Luo J. Single-cell and spatial transcriptomics reveals that PTPRG activates the m 6A methyltransferase VIRMA to block mitophagy-mediated neuronal death in Alzheimer's disease. Pharmacol Res 2024; 201:107098. [PMID: 38325728 DOI: 10.1016/j.phrs.2024.107098] [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: 11/10/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Neuronal death is one of the key pathologies in Alzheimer's disease (AD). How neuronal death begins in AD is far from clear, so clarifying this process may help develop effective therapies. This study collected single-cell RNA sequencing data of 85 AD samples and 83 control samples, covering the prefrontal cortex, internal olfactory cortex, superior parietal lobe, superior frontal gyrus, caudal internal olfactory cortex, somatosensory cortex, hippocampus, superior frontal cortex and peripheral blood mononuclear cells. Additionally, spatial transcriptomic data of coronal sections from 6 AppNL-G-F AD mice and 6 control C57Bl/6 J mice were acquired. The main single-cell and spatial transcriptomics results were experimentally validated in wild type and 5 × FAD mice. We found that the microglia subpopulation Mic_PTPRG can communicate with specific types of neurons (especially excitatory ExNeu_PRKN_VIRMA and inhibitory InNeu_PRKN_VIRMA neuronal subpopulations) and cause them to express PTPRG during AD progression. Within neurons, PTPRG binds and upregulates the m6A methyltransferase VIRMA, thus inhibiting translation of PRKN mRNA to prevent the clearance of damaged mitochondria in neurons through suppressing mitophagy. As the disease progresses, the energy and nutrient metabolic pathways in neurons are reprogrammed, leading to their death. Consistently, we determined that PTPTRG can physically interact with VIRMA in mouse brains and PRKN is significantly upregulated in 5 × FAD mouse brain. Altogether, our findings demonstrate that PTPRG activates the m6A methyltransferase VIRMA to block mitophagy-mediated neuronal death in AD, which is a potential pathway, through which microglia and neuronal PTPRG modify neuronal connections in the brain during AD progression.
Collapse
Affiliation(s)
- Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China.
| | - Xiaohua Huang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Yating Lan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Jieqiong Xie
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Qi Huang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Jingyi Zeng
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Chun Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Zifei Pei
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Cuihua Zou
- Guangxi Medical University Cancer Hospital, Nanning 530022, Guangxi, China.
| | - Yingwei Mao
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA.
| | - Jiefeng Luo
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China.
| |
Collapse
|
16
|
Wang C, Cui X, Dong Z, Liu Y, Xia P, Wang X, Zhang Z, Yu S, Wu S, Liu H, Zong S, Lu Z. Attenuated memory impairment and neuroinflammation in Alzheimer's disease by aucubin via the inhibition of ERK-FOS axis. Int Immunopharmacol 2024; 126:111312. [PMID: 38043266 DOI: 10.1016/j.intimp.2023.111312] [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: 08/23/2023] [Revised: 11/11/2023] [Accepted: 11/26/2023] [Indexed: 12/05/2023]
Abstract
Alzheimer's disease (AD) is a degenerative illness accompanied by cognitive and memory loss. In addition to the widely accepted, convincing amyloid cascade hypothesis, the activation of glial cells and neuroinflammation, especially the microglia-mediated neuroinflammation, has an essential role in the development and progression of AD. Therefore, the anti-inflammatory treatment is becoming a promising therapeutic strategy. Aucubin (Au) is a natural product derived from many plants with anti-inflammatory and antioxidant activities. Up to now, no research has been conducted to investigate the anti-inflammatory effects of Au and its neuroprotective quality on AD and the potential molecular mechanisms of its medical roles. In our study, the results of network pharmacology revealed the potential therapeutic effect of Au on AD. The results of studies in vivo showed that Au improved the behaviors, counteracted cognitive and memory deficits, and ameliorated AD-like pathological features of the mouse brain, e.g., the deposition of Aβ plaques, neuronal damage, and inflammatory responses induced by glial cell overactivation, in APP/PS1 mice. The transcriptome sequencing further confirmed that the pathological symptoms of AD could be reversed by inhibiting the ERK/FOS axis to alleviate the inflammatory response. The in vitro experiments revealed that Au suppressed the BV2 cell activation, inhibited the phosphorylation of ERK1/2 and the expression of c-FOS, and reduced the LPS-induced inflammatory mediator production by BV2 cells and primary astrocytes. Our study suggested that Au exerted its neuroprotective effects by inhibiting the inflammatory responses, which could be a promising treatment of AD.
Collapse
Affiliation(s)
- Cuicui Wang
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaolin Cui
- School of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhenfang Dong
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yingchao Liu
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Pengcheng Xia
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xueying Wang
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhi Zhang
- School of Medicine, Shandong University, Jinan, Shandong, China
| | - Shuyi Yu
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shuang Wu
- School of Medicine, Shandong University, Jinan, Shandong, China
| | - Huan Liu
- School of Medicine, Shandong University, Jinan, Shandong, China
| | - Shuai Zong
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Zhiming Lu
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
17
|
Geng Y, Wang Z, Xu X, Sun X, Dong X, Luo Y, Sun X. Extensive therapeutic effects, underlying molecular mechanisms and disease treatment prediction of Metformin: a systematic review. Transl Res 2024; 263:73-92. [PMID: 37567440 DOI: 10.1016/j.trsl.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
Metformin (Met), a first-line management for type 2 diabetes mellitus, has been expansively employed and studied with results indicating its therapeutic potential extending beyond glycemic control. Beyond its established role, this therapeutic drug demonstrates a broad spectrum of action encompassing over 60 disorders, encompassing metabolic conditions, inflammatory disorders, carcinomas, cardiovascular diseases, and cerebrovascular pathologies. There is clear evidence of Met's action targeting specific nodes in the molecular pathways of these diseases and, intriguingly, interactions with the intestinal microbiota and epigenetic processes have been explored. Furthermore, novel Met derivatives with structural modifications tailored to diverse diseases have been synthesized and assessed. This manuscript proffers a comprehensive thematic review of the diseases amenable to Met treatment, elucidates their molecular mechanisms, and employs informatics technology to prospect future therapeutic applications of Met. These data and insights gleaned considerably contribute to enriching our understanding and appreciation of Met's far-reaching clinical potential and therapeutic applicability.
Collapse
Affiliation(s)
- Yifei Geng
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Zhen Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Xiaoyu Xu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Xiao Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Xi Dong
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China.
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China.
| |
Collapse
|
18
|
Chen X, Song Y, Song W, Han J, Cao H, Xu X, Li S, Fu Y, Ding C, Lin F, Shi Y, Li J. Multi-omics reveal neuroprotection of Acer truncatum Bunge Seed extract on hypoxic-ischemia encephalopathy rats under high-altitude. Commun Biol 2023; 6:1001. [PMID: 37783835 PMCID: PMC10545756 DOI: 10.1038/s42003-023-05341-9] [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: 04/27/2023] [Accepted: 09/11/2023] [Indexed: 10/04/2023] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) at high-altitudes leads to neonatal mortality and long-term neurological complications without effective treatment. Acer truncatum Bunge Seed extract (ASO) is reported to have effect on cognitive improvement, but its molecular mechanisms on HIE are unclear. In this study, ASO administration contributed to reduced neuronal cell edema and improved motor ability in HIE rats at a simulated 4500-meter altitude. Transcriptomics and WGCNA analysis showed genes associated with lipid biosynthesis, redox homeostasis, neuronal growth, and synaptic plasticity regulated in the ASO group. Targeted and untargeted-lipidomics revealed decreased free fatty acids and increased phospholipids with favorable ω-3/ω-6/ω-9 fatty acid ratios, as well as reduced oxidized glycerophospholipids (OxGPs) in the ASO group. Combining multi-omics analysis demonstrated FA to FA-CoA, phospholipids metabolism, and lipid peroxidation were regulated by ASO treatment. Our results illuminated preliminary metabolism mechanism of ASO ingesting in rats, implying ASO administration as potential intervention strategy for HIE under high-altitude.
Collapse
Affiliation(s)
- Xianyang Chen
- Bao Feng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Yige Song
- Bao Feng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Wangting Song
- Bao Feng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Jiarui Han
- Bao Feng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Hongli Cao
- Department of Respiratory, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Xiao Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Plateau Medical Research Center of China Medical University, Shenyang, China
| | - Shujia Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Plateau Medical Research Center of China Medical University, Shenyang, China
| | - Yanmin Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Plateau Medical Research Center of China Medical University, Shenyang, China
| | - Chunguang Ding
- National Center for Occupational Safety and Health, Beijing, China
| | - Feng Lin
- Department of Neurology, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, Fujian, China
| | - Yuan Shi
- Department of Neonatology, Children's Hospital Affiliated Chongqing Medical University, Chongqing, China
| | - Jiujun Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Plateau Medical Research Center of China Medical University, Shenyang, China.
| |
Collapse
|
19
|
Xue Z, Ye L, Ge J, Lan Z, Zou X, Mao C, Bao X, Yu L, Xu Y, Zhu X. Wwl70-induced ABHD6 inhibition attenuates memory deficits and pathological phenotypes in APPswe/PS1dE9 mice. Pharmacol Res 2023; 194:106864. [PMID: 37480972 DOI: 10.1016/j.phrs.2023.106864] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 07/04/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Synaptic dysfunction plays a crucial role in the pathogenesis of Alzheimer's disease (AD). α/β-hydrolase domain-containing 6 (ABHD6) contributes to synaptic dysfunctions, and ABHD6 inhibition has shown potential therapeutic value in neurological disorders. However, the role of ABHD6 in AD has not been fully defined. In this study, we demonstrated that adeno-associated virus (AAV) mediated shRNA targeting ABHD6 in hippocampal neurons attenuated synaptic dysfunction and memory impairment of APPswe/PS1dE9 (APP/PS1) mice, while it didn't affect the amyloid-beta (Aβ) levels and neuroinflammation in the brains. In addition, intraperitoneal injection of wwl70, a specific inhibitor of ABHD6, improved synaptic plasticity and memory function in APP/PS1 mice, which might attribute to the activation of endogenous cannabinoid signaling. Furthermore, wwl70 significantly decreased the Aβ levels and neuroinflammation in the hippocampus of AD mice, and enhanced Aβ phagocytized by microglia. In conclusion, for the first time our data have shown that ABHD6 inhibition might be a promising strategy for AD treatment, and wwl70 is a potential candidate for AD drug development pipeline.
Collapse
Affiliation(s)
- Zhiwei Xue
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Lei Ye
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Jianwei Ge
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Zhen Lan
- Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinxin Zou
- Department of Neurology, Drum Tower Hospital of Xuzhou Medical University, Nanjing, Jiangsu, China
| | - Chenglu Mao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Linjie Yu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China; Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurology, Drum Tower Hospital of Xuzhou Medical University, Nanjing, Jiangsu, China
| | - Xiaolei Zhu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China; Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
20
|
Shen ZJ, Fu YB, Hou JL, Lin LN, Wang XY, Li CY, Yang YX. Integrating network pharmacology, UPLC-Q-TOF-MS and molecular docking to investigate the effect and mechanism of Chuanxiong Renshen decoction against Alzheimer's disease. Chin Med 2022; 17:143. [PMID: 36566207 PMCID: PMC9789652 DOI: 10.1186/s13020-022-00698-1] [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: 09/01/2022] [Accepted: 12/08/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND AIM Chuanxiong Renshen decoction (CRD) is a traditional Chinese medicine compound used to treat Alzheimer's disease (AD). However, the effects and active ingredients of CRD and its mechanism have not been clarified. We aimed to determine the neuroprotective effects of CRD in a triple-transgenic mouse model of AD (3 × Tg-AD) and investigate the possible active ingredients and their mechanisms. METHODS Morris water maze (MWM) tests were used to determine the protective effect of CRD on learning and memory ability. Afterward, we used brain tissue staining, immunofluorescent staining and western blotting to detect the neuroprotective effects of CRD. Ultraperformance liquid-chromatography-quadrupole-time-of-flight tandem mass spectrometry (UPLC-Q-TOF-MS) was applied to determine the ingredients of CRD, and the potential AD targets were obtained from DisGeNET and the GeneCards database. The protein‒protein interaction (PPI) network was built with the additional use of STRING 11.0. Metascape was used in the pathway enrichment analysis. Discovery Studio 2016 (DS) software was used to analyze the binding ability of CRD and AD-related genes. Finally, we verified the regulatory effect of CRD on the predicted core targets EGFR and CASP3 by western blotting. RESULTS Our study indicated that CRD can significantly improve learning and memory, reduce the expression of Aβ and protect neurons. A total of 95 ingredients were identified in the CRD. Then, 25 ingredients were identified in serum, and 5 ingredients were identified in the brain tissue homogenate. PPI network analysis identified CASP3, EGFR, APP, CNR1, HIF1A, PTGS2 and MTOR as hub targets. KEGG and GO analyses revealed that the TNF signaling pathway and MAPK signaling pathway were enriched in multiple targets. The results of molecular docking proved that the binding of the ingredients with potential key targets was excellent. The western blotting results showed that CRD could significantly reduce the expression of CASP3 and EGFR in the hippocampus of 3 × Tg-AD mice. Combined with literature analysis, we assumed the neuroprotective effect of CRD on AD may occur through regulation of the MAPK signaling pathway. CONCLUSION CRD significantly alleviated injury in 3 × Tg-AD mice. The possible active ingredients are ferulic acid, rutin, ginsenoside Rg1 and panaxydol. The therapeutic effect of CRD on AD is achieved through the downregulation of CASP3 and EGFR. The neuroprotective effect of CRD on AD may occur through regulation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Zhuo Jun Shen
- grid.506977.a0000 0004 1757 7957School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Yun Bo Fu
- grid.268505.c0000 0000 8744 8924Department of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jin Ling Hou
- grid.506977.a0000 0004 1757 7957School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Lu Ning Lin
- grid.268505.c0000 0000 8744 8924Department of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiao Yan Wang
- grid.506977.a0000 0004 1757 7957School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Chang Yu Li
- grid.268505.c0000 0000 8744 8924Department of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuan Xiao Yang
- grid.506977.a0000 0004 1757 7957School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|