1
|
Liu Y, Chen H, Chen K, Shang Q, Lin F, Zhang P, Zhang Y, Qin W, Liu H, Qiu W, Chen X, Gong Y, Jiang Y, Zhang Y, He J, Zhao W, Ren H, Shen G, Jiang X. Luteolin alleviates estrogen deficiency-induced muscle atrophy via targeting SLC7A11-mediated ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156799. [PMID: 40300261 DOI: 10.1016/j.phymed.2025.156799] [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: 03/25/2025] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Skeletal muscle atrophy, which is a debilitating condition exacerbated by estrogen deficiency, lacks effective therapeutic interventions. Although ferroptosis (an iron-dependent form of cell death driven by lipid peroxidation) has emerged as a contributor to muscle degeneration, its regulatory mechanisms remain poorly defined. In this study, we identified luteolin, which is a natural flavonoid, as a potent inhibitor of ferroptosis that mitigates estrogen deficiency-induced muscle atrophy by targeting SLC7A11. PURPOSE The aim of this study was to investigate the role of ferroptosis in the anti-muscle atrophy effects of luteolin. METHODS Via database screening, luteolin was identified as a potential drug for improving muscle atrophy, and the promotion of C2C12 myogenic differentiation by luteolin was detected by using immunofluorescence (IF), quantitative reverse transcription PCR (RT-qPCR) and western blot (WB). The mechanism of luteolin-mediated ferroptosis in muscle atrophy was confirmed by RNA-seq, transmission electron microscopy (TEM), and GSH, MDA, SOD and Fe2+ assays. Molecular docking, molecular dynamics simulation, surface plasmon resonance (SPR), cellular thermal shift assay (CETSA), drug affinity responsive target stability (DARTS) and siRNA-mediated gene knockout were applied to validate the notion that the mechanism of luteolin treatment of muscle atrophy involves target binding to SLC7A11. In addition, this study confirmed the role of luteolin in ameliorating muscle atrophy via the modulation of the SLC7A11-mediated ferroptosis pathway in vivo. Finally, the effect of luteolin on the myogenic differentiation of HsKMCs was investigated. RESULTS Luteolin promotes myogenic differentiation and significantly inhibits myotube atrophy, with the main mechanism of these effects involving the direct binding of luteolin to the SLC7A11 protein to inhibit the occurrence of ferroptosis. We confirmed that luteolin can inhibit ferroptosis in muscle tissue and improve the loss of muscle mass and strength due to muscle atrophy in vivo. In addition, luteolin significantly inhibited myotube atrophy in HsKMCs and promoted their myogenic differentiation by modulating the SLC7A11-mediated ferroptosis. CONCLUSIONS Our findings demonstrate that luteolin regulates myogenesis and prevents muscle atrophy through binding to SLC7A11 and subsequently inhibiting ferroptosis. This study elucidates the critical role of the SLC7A11-ferroptosis axis in preserving muscle physiology during atrophy, while identifying luteolin as a therapeutic agent capable of targeting SLC7A11 to suppress ferroptosis and alleviate muscle atrophy.
Collapse
Affiliation(s)
- Yu Liu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Honglin Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Kun Chen
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, PR China; Guangzhou Medical University, Guangzhou 511436, PR China
| | - Qi Shang
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, PR China; Guangzhou Medical University, Guangzhou 511436, PR China
| | - Feng Lin
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Peng Zhang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - You Zhang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Weicheng Qin
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Huiwen Liu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Weiyu Qiu
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, PR China; Guangzhou Medical University, Guangzhou 511436, PR China
| | - Xingda Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yan Gong
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yixuan Jiang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yuzhuo Zhang
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, PR China; Guangzhou Medical University, Guangzhou 511436, PR China
| | - Jiahui He
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510130, PR China
| | - Wenhua Zhao
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, PR China; Guangzhou Medical University, Guangzhou 511436, PR China
| | - Hui Ren
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, PR China; Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Gengyang Shen
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, PR China; Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Xiaobing Jiang
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, PR China; Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
2
|
Tan H, Fu X, Yang R, Tang J, Zeng S, Liu Z, Zhu X, Zhang X, Xie L, Wu D. Dual targeting of FSP1 and xCT: Potential mechanism of anthocyanins in alleviating neuronal ferroptosis in vascular dementia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156608. [PMID: 40334339 DOI: 10.1016/j.phymed.2025.156608] [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: 12/16/2024] [Revised: 02/12/2025] [Accepted: 03/02/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND VaD, the second most prevalent type of dementia in the elderly following Alzheimer's disease, is marked by significant cognitive and motor deficits, with few effective treatment options currently available. Ferroptosis, a type of regulated cell death driven by iron-mediated lipid peroxidation, has recently emerged as a key pathological mechanism in the development of VaD. Ferroptosis drives neuronal damage in VaD, making it a promising therapeutic target to reduce neuronal death and preserve cognitive function. ACN, a group of polyphenolic compounds recognized for their strong antioxidant properties, have demonstrated potential in reducing ferroptosis and alleviating neuronal damage. OBJECTIVE The aim of this study was to explore the neuroprotective effects of ACN in reducing ferroptosis and mitigating cognitive impairments associated with VaD, focusing on the dual modulation of the FSP1 and xCT/GPX4 pathways. This novel dual-target approach provides an innovative strategy to reduce neuronal damage and oxidative stress in VaD. METHODS A combination of in vitro and in vivo experiments was conducted to assess the protective effects and underlying mechanisms of ACN in mitigating ferroptosis associated with VaD. In vitro, a neurotoxicity model was established by inducing PC12 cells with Glu. Cell viability was determined using the CCK-8 assay, and various markers, including ROS levels, MDA, LPO, and GSH levels, were measured to evaluate the protective effects of ACN. Additionally, the expression of ferroptosis-related proteins, such as FSP1, xCT, and GPX4, was analyzed through Western blotting, RT-qPCR, and immunofluorescence. In vivo, a VaD rat model was established by performing bilateral common carotid artery occlusion (2-VO). The rats were divided into four groups: control, model, ACN-treated (with varying doses), and ALA-treated (positive control). The intervention lasted for 28 days. Cognitive functions were assessed using the Morris water maze and novel object recognition tests. Histological analyses, including HE staining and Nissl staining, were carried out to examine neuronal pathology. Moreover, electron microscopy was employed to evaluate mitochondrial ultrastructure integrity. Brain levels of iron, lipid peroxidation markers, and the expression of FSP1, xCT, and GPX4 were measured to elucidate the molecular mechanisms underlying the observed effects. RESULTS Systematic in vitro and in vivo experiments demonstrated the significant neuroprotective effects of ACN against ferroptosis associated with VaD. In the Glu-induced PC12 cell model, ACN significantly improved cell viability, reduced ROS levels, restored GSH levels, and decreased the accumulation of MDA and LPO. Notably, ACN upregulated the expression of key ferroptosis-suppressing proteins, FSP1, xCT, and GPX4, through dual activation of these pathways, highlighting its powerful protective role against oxidative stress and ferroptosis. In the 2-VO VaD rat model, high-dose ACN significantly improved cognitive function, as shown by reduced escape latency in the Morris water maze and increased platform crossings. Moreover, ACN treatment enhanced the discrimination index in the novel object recognition test, suggesting improved learning and memory. Histopathological analyses revealed that ACN significantly alleviated neuronal disorganization, increased Nissl body counts, and restored mitochondrial integrity, with reduced swelling, rupture, and vacuolation observed under electron microscopy. CONCLUSION ACN exerts significant neuroprotective effects in VaD by dual regulation of the FSP1 and xCT/GPX4 pathways, effectively inhibiting ferroptosis and alleviating oxidative stress. This "dual-target" mechanism not only expands the current understanding of ACN's neuroprotective effects but also emphasizes its unique role in inhibiting ferroptosis. Overall, this study provides experimental evidence supporting the potential use of ACN in treating ferroptosis-related neurodegenerative diseases and highlights its promising prospects for clinical application.
Collapse
Affiliation(s)
- Huizhong Tan
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Institute of Innovation and Applied Research, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinying Fu
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Institute of Innovation and Applied Research, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Renyi Yang
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Institute of Innovation and Applied Research, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Tang
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Institute of Innovation and Applied Research, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shanshan Zeng
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Institute of Innovation and Applied Research, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhuxuan Liu
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Institute of Innovation and Applied Research, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinhua Zhu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiuli Zhang
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Institute of Innovation and Applied Research, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| | - Le Xie
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Institute of Innovation and Applied Research, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| | - Dahua Wu
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Institute of Innovation and Applied Research, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
3
|
Peng W, Liang J, Qian X, Li M, Nie M, Chen B. IGF2BP1/AIFM2 axis regulates ferroptosis and glycolysis to drive hepatocellular carcinoma progression. Cell Signal 2025; 130:111660. [PMID: 39971223 DOI: 10.1016/j.cellsig.2025.111660] [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: 01/24/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is aggressive liver tumor that is the third leading cause of cancer death. Ferroptosis and glycolysis play key roles in HCC progression. Apoptosis-inducing factor mitochondria-associated 2 (AIFM2) in involved in regulating ferroptosis and glycolysis in cancers, but its role in HCC remains unclear. This research explored the function of AIFM2 in HCC. METHODS AIFM2 expression in HCC tissues was evaluated using the UALCAN and GEPIA databases, as well as RT-qPCR. Kaplan-Meier survival analysis analyzed the correlation between AIFM2 and the prognosis of HCC patients. EdU and transwell assays were utilized to examine HCC cell proliferation, migration, and invasion. Ferroptosis markers were analyzed by measuring iron levels, ROS production (DCFH-DA assay), and oxidative stress indicators (SOD, MDA, and GSH). Glycolytic activity was assessed through glucose uptake, lactate production, and ATP levels. m6A modification on AIFM2 mRNA was confirmed by MeRIP assay, and mRNA stability was evaluated with Actinomycin D treatment. Tumor growth and metastasis were studied in xenograft and lung metastasis models. RESULTS UALCAN analysis showed that AIFM2 was significantly upregulated in HCC tissues, which correlated with poor survival rates of HCC patients. IGF2BP1 was also highly expressed in HCC tissues and positively correlated with AIFM2 levels in HCC tissues. Functionally, AIFM2 knockdown suppressed glycolysis and enhanced ferroptosis, while its overexpression had opposite effects. IGF2BP1 was found to stabilize AIFM2 mRNA via m6A modification, promoting AIFM2 expression. IGF2BP1 knockdown reduced glycolysis, proliferation, and invasion while promoting ferroptosis, while AIFM2 overexpression could reverse this effect. In vivo, IGF2BP1 or AIFM2 silencing significantly suppressed tumor growth and metastasis. CONCLUSION IGF2BP1 stabilized AIFM2 mRNA to regulate ferroptosis and glycolysis and promoted HCC progression.
Collapse
Affiliation(s)
- Wei Peng
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Jie Liang
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Xuanlv Qian
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Mingwang Li
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Ming Nie
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Bin Chen
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China.
| |
Collapse
|
4
|
Mai L, Liu J, Wu H, Wang H, Lin Z, Rao S, Sun W, Tan A, Lin Y, Chen B. Enhanced inhibition of neuronal ferroptosis and regulation of microglial polarization with multifunctional traditional Chinese medicine active ingredients-based selenium nanoparticles for treating spinal cord injury. Mater Today Bio 2025; 32:101758. [PMID: 40270889 PMCID: PMC12017924 DOI: 10.1016/j.mtbio.2025.101758] [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: 01/01/2025] [Revised: 03/23/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that results in the loss of sensory and motor functions. The complex pathogenesis of SCI contributes to the limited availability of effective therapies. Two major factors exacerbating secondary injury in SCI are neuronal ferroptosis and microglial inflammatory polarization. Tanshinone IIA (TSIIA) has demonstrated a significant anti-ferroptosis effect by inhibiting lipid peroxidation, while tetramethylpyrazine (TMP) exhibits remarkable anti-inflammatory properties by promoting the shift of microglial polarization from the M1 to the M2 phenotype. However, most drugs currently under development primarily target a single aspect of this multifaceted condition, which is insufficient for comprehensive treatment. Selenium nanoparticles have emerged as a promising therapeutic strategy due to their ability to encapsulate various agents, effectively targeting diverse pathophysiological mechanisms while offering favorable water solubility and low toxicity. In this study, we developed a novel nanocarrier functionalized with astragalus polysaccharide (APS) and loaded with TSIIA and TMP. Results from both in vitro and in vivo studies indicate that TSIIA/TMP/APS@Se NPs possess anti-ferroptosis properties and can regulate microglial polarization, potentially enhancing functional recovery following SCI. In summary, this study presents a promising alternative strategy for treating SCI, highlighting its significant potential for future clinical applications.
Collapse
Affiliation(s)
- Luoqi Mai
- Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Jinggong Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Huimei Wu
- Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Dermatology Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Hongshen Wang
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Zhidong Lin
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Siyuan Rao
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Wenxi Sun
- Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Aowei Tan
- Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yongpeng Lin
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Bolai Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Orthopedics Department, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| |
Collapse
|
5
|
Chen Y, Wang Q, Bian S, Dong J, Xiong J, Le J. Exploration of the mechanism of Polyphyllin I against hepatocellular carcinoma based on network pharmacology, molecular docking and experimental validation. Discov Oncol 2025; 16:941. [PMID: 40434621 PMCID: PMC12120097 DOI: 10.1007/s12672-025-02341-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 04/08/2025] [Indexed: 05/29/2025] Open
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide. Targeted therapies hold promise for HCC treatment, and understanding the molecular mechanisms of action is crucial for developing novel therapeutic strategies. Polyphyllin I, a natural compound with known antitumor activity, represents a potential therapeutic candidate. METHODS This study employed a network pharmacology approach to investigate the anti-HCC effects of Polyphyllin I and its underlying mechanisms. Drug and disease related targets were identified and intersected to construct Components-Gene Symbols-Disease and Protein-Protein Interaction networks. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. Molecular docking simulations were conducted to explore the interactions between Polyphyllin I and key pathway proteins (VEGF-C and β-catenin). Finally, in vitro and in vivo experiments validated the anti-HCC effects and underlying mechanisms of Polyphyllin I. RESULTS Network pharmacology analysis revealed that Polyphyllin I targets multiple genes and pathways implicated in HCC development and progression. GO and KEGG analyses identified significant enrichment of pathways related to cell proliferation, apoptosis and angiogenesis, including VEGF and the Wnt/β-catenin signaling pathways. Molecular docking simulations demonstrated strong binding affinities between Polyphyllin I and VEGF-C and β-catenin. In vitro and in vivo experiments confirmed that Polyphyllin I effectively inhibits HCC cell proliferation, induces apoptosis, and suppresses angiogenesis, potentially by modulating the VEGF-C and Wnt/β-catenin signaling pathways. CONCLUSIONS The study provides compelling evidence for the antitumor activity of Polyphyllin I in HCC and elucidates its possible molecular mechanisms, suggesting that Polyphyllin I holds great potential as a therapeutic agent for HCC.
Collapse
Affiliation(s)
- Yilong Chen
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Qiuying Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Shuixiu Bian
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jing Dong
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jie Xiong
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Jiamei Le
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
| |
Collapse
|
6
|
Zhao H, Zhang H, Shi J, Liu Y, Yu J, Yang Y, Weng J, Song Z, Zhou R, Min H, Yao J, Wang M, Zhang Z. Wuwei Shaji powder alleviates OVA-induced allergic asthma by protecting bronchial epithelial cells from ferroptosis via the S-sulfhydration of Keap1. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119649. [PMID: 40222689 DOI: 10.1016/j.jep.2025.119649] [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: 01/08/2025] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 04/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Asthma is a chronic inflammatory airway disease. Current treatments have limited efficacy and often cause severe side effects. Wuwei Shaji Powder (WSP), a traditional Mongolian remedy, is used for treating chronic pulmonary diseases, but its efficacy against asthma and underlying mechanisms are still unclear. AIM OF THE STUDY To investigate the therapeutic effect of WSP on asthma and elucidate its molecular mechanisms. MATERIALS AND METHODS An ovalbumin (OVA)-induced allergic asthma model was established in rats. Ferroptosis or apoptosis was induced in BEAS-2B cells using Erastin or CdCl2. Various techniques including histopathological staining, ELISA, Western blot, flow cytometry, and transmission electron microscopy were employed to assess WSP's effects and mechanisms. RESULTS WSP alleviated OVA-induced allergic asthma in rats without the immunosuppressive side effects observed with dexamethasone. WSP suppressed ferroptosis in bronchial epithelial cells both in vivo and in vitro. It reduced thiol- and sulfonic-based oxidative stress through Keap1 S-sulfhydration, disrupted the Keap1-Nrf2 interaction, and promoted Nrf2 nuclear translocation. Notably, we discovered that CdCl2 can induce ferroptosis in BEAS-2B cells, and WSP prevented both ferroptosis and apoptosis in these cells. CONCLUSION WSP alleviates OVA-induced allergic asthma by protecting bronchial epithelial cells from ferroptosis via S-sulfhydration of Keap1, providing new insights for its clinical application.
Collapse
Affiliation(s)
- Huimei Zhao
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Haiyan Zhang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jianyu Shi
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yanru Liu
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jingao Yu
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yuangui Yang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jingyu Weng
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Zhongxing Song
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Rui Zhou
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Hong Min
- NMPA Key Laboratory for Testing Technology of Pharmaceutical Microbiology, Shaanxi Institute for Food and Drug Control, Xi'an, China
| | - Jian Yao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Mei Wang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China; Wangjing Hospital of China Academy of Traditional Chinese Medicine, Beijing, 100102, China.
| | - Zhen Zhang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| |
Collapse
|
7
|
Wang C, Shi CH, Bai HY, Lu J, Hu HT, Sun YM, Gao H, An H, Lu JH, Zhao HJ, Zhu ZH. Astragali radix - Curcumae rhizoma herb pair suppresses hepatocellular carcinoma through EGFR/AKT/mTOR pathway and induces lipid peroxidation-related ferroptosis via HIF-1α/HO-1/GPX4 axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119912. [PMID: 40316156 DOI: 10.1016/j.jep.2025.119912] [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: 03/27/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Astragali Radix - Curcumae Rhizoma herb pair (ACHP) originated from the famous traditional Chinese medicine text "YiXueZhongZhongCanXiLu", in which the two herbs were paired to form Chinese herbal compounds commonly used clinically for digestive system tumors, such as hepatocellular carcinoma (HCC). Although ACHP has been inherited for thousands of years in China, its mechanism against HCC remains unclear. AIM OF THE STUDY The study aims to evaluate the effect and explore the mechanism of ACHP against HCC. METHODS The efficacy and safety of ACHP against HCC in vivo were evaluated by tumor volume, organ index, H&E staining, hepatic and renal factors. The serum metabolites of ACHP were identified by UPLC-Q-TOF-MS/MS. The key targets and potential mechanisms of ACHP against HCC were screened by transcriptomics, network pharmacology and molecular docking. The effect and induction of ferroptosis of ACHP-containing serum on HCC in vitro was evaluated by MTT, colony formation assay and specific detection kits. The expression of ferroptosis-related proteins and pathways in vivo was detected by immunohistochemistry. RESULTS ACHP significantly inhibited tumor proliferation compared to the two herbs used separately, and showed a favorable safety profile. A total of 75 serum metabolites were identified in both positive and negative ion modes. Transcriptomics results revealed that ferroptosis played a key role in the anti-HCC process of ACHP. Network pharmacology and molecular docking results suggested that the anti-HCC effect of ACHP may be related to EGFR/AKT/mTOR pathway and HIF-1α/HO-1/GPX4 axis. In vitro and in vivo experiments further demonstrated that ACHP suppressed oncogenic signaling via the EGFR/AKT/mTOR pathway while inducing lipid peroxidation-related ferroptosis through HIF-1α/HO-1/GPX4 axis, thereby inhibiting HepG2 cells proliferation and HCC mice tumor growth. CONCLUSION ACHP exerts its effects by suppressing oncogenic signaling through the EGFR/AKT/mTOR pathway and inducing lipid peroxidation-related ferroptosis in HCC via the HIF-1α/HO-1/GPX4 axis. This systematic investigation establishes a coherent pharmacological chain from compound identification to mechanism verification, highlighting ACHP's therapeutic potential as a ferroptosis inducer targeting oncogenic signaling networks in HCC.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Animals
- Humans
- Proto-Oncogene Proteins c-akt/metabolism
- Ferroptosis/drug effects
- ErbB Receptors/metabolism
- TOR Serine-Threonine Kinases/metabolism
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Mice
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lipid Peroxidation/drug effects
- Male
- Mice, Inbred BALB C
- Signal Transduction/drug effects
- Mice, Nude
- Antineoplastic Agents, Phytogenic/pharmacology
- Cell Line, Tumor
- Hep G2 Cells
- Rhizome
- Xenograft Model Antitumor Assays
- Astragalus propinquus
Collapse
Affiliation(s)
- Chen Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Chen-Hao Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Hao-Yang Bai
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Jun Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Hong-Tao Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Yu-Mei Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Hang Gao
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Hai An
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Jia-Hui Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Hua-Jun Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Zhi-Hui Zhu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
8
|
Zhang H, Yuan F, Zhao N, Tang W, Zhao P, Liu C, Chen S, Hou X, Xia C, Chu J. Nanoparticle-mediated SIRT1 inhibition suppresses M2 macrophage polarization and hepatocarcinogenesis in chronic hepatitis B. J Nanobiotechnology 2025; 23:385. [PMID: 40426198 PMCID: PMC12117705 DOI: 10.1186/s12951-025-03447-2] [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: 02/04/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a major complication of chronic hepatitis B (CHB), with macrophage M2 polarization playing a critical role in shaping the tumor-promoting hepatic immune microenvironment. Sirtuin 1 (SIRT1) has been implicated in immune modulation and liver carcinogenesis. This study investigates the potential of Mimetic Nanoparticles (MNPs) for delivering SIRT1 inhibitors to regulate macrophage polarization and remodel the hepatic immune microenvironment, aiming to prevent HCC development post-CHB. A transgenic mouse model of CHB was established, and RNA sequencing (RNA-seq) and proteomics analyses revealed significant dysregulation of genes associated with M2 macrophage polarization, particularly SIRT1. Functional enrichment analysis highlighted key pathways, including PI3K-Akt and NF-κB, that contribute to CHB-driven immune alterations. Synthesized and characterized MNPs successfully delivered SIRT1 inhibitors, effectively inhibiting M2 macrophage polarization in vitro. In vivo administration of MNPs-SIRT1-FN significantly reduced M2 macrophage infiltration and suppressed tumor growth. These findings suggest that nanoparticle-mediated SIRT1 inhibition is a promising strategy for immunomodulation and HCC prevention in CHB patients. This study provides novel insights into nanoparticle-based immunotherapy for CHB-related HCC and highlights a potential therapeutic avenue for liver cancer prevention.
Collapse
Affiliation(s)
- He Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Feng Yuan
- Laboratory of Inflammation and Vaccines, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Nan Zhao
- Institute of Clinical Medicine, National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Wenqiang Tang
- Institute of Animal Science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa, 850009, China
| | - Pengwei Zhao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Chunfa Liu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China
| | - Shan Chen
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China
| | - Xiaolin Hou
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China.
| | - Changyou Xia
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China.
| | - Jun Chu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China.
- Institute of Animal Science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa, 850009, China.
| |
Collapse
|
9
|
Zhou J, Shi Y, Jian Y, Li Y, Yu W, Mu W, Ge Y. Identification of key ferroptosis genes in hepatocellular carcinoma and type 2 diabetes mellitus through bioinformatics analysis. Discov Oncol 2025; 16:916. [PMID: 40413683 DOI: 10.1007/s12672-025-02758-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Ferroptosis is a programmed cell death mode associated with iron metabolism, with accumulation of intracellular lipid peroxides, which is closely related to the occurrence and development of multiple diseases, including type 2 diabetes mellitus (T2DM) and hepatocellular carcinoma (HCC). T2DM is a chronic metabolic disorder characterized by a combination of impaired insulin sensitivity and insufficient insulin production, frequently accompanied by obesity and fatty liver, which increases the risk of developing HCC. To explore the complex interactions between ferritin deposition, T2DM, and HCC, we performed bioinformatics analysis on publicly available gene expression data and identified 23 differentially expressed genes (DEGs) that are commonly expressed in both T2DM and HCC. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that these DEGs are primarily enriched in fatty acid metabolism and ferroptosis pathways. The weighted gene co-expression network analysis (WGCNA) identified 6 key genes associated with the pathogenesis of both diseases. Taking the intersection of DEGs and iron deposition-related genes, we identified ACSL4 as a key ferroptosis gene involved in the co-morbidity of T2DM and HCC. To validate the bioinformatics findings, we assessed the expression of ACSL4 using Receiver operating characteristic (ROC) curve analysis, which revealed an Area Under the Curve (AUC) of 0.886 for HCC and 0.745 for T2DM. Additionally, an insulin resistance model was established in HepG2 cells by treatment with 350 µM palmitic acid (PA), resulting in significant changes in cell morphology. Oil Red O staining showed a marked increase in lipid accumulation. RT-PCR analysis further confirmed the significant alteration in ACSL4 gene expression. In conclusion, this study is the first to integrate bioinformatics tools to investigate the potential mechanistic links between iron metabolism and the comorbidity of T2DM and HCC, uncovering a novel pathogenic pathway. These findings provide new directions for drug development and therapeutic strategies in the future.
Collapse
Affiliation(s)
- Jinjin Zhou
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yage Shi
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yulun Jian
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuhan Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenya Yu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Mu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yang Ge
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
10
|
Liu WM, Li XB. East meets West: The winning combination against BRAF V600E metastatic colorectal cancer. World J Clin Oncol 2025; 16:102223. [DOI: 10.5306/wjco.v16.i5.102223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 05/19/2025] Open
Abstract
Metastatic colorectal cancer (mCRC) patients with BRAF V600E mutation have a poor prognosis despite the implementation of multiple treatment strategies. The integration of traditional Chinese medicine with Western medicine in treating BRAF mutant mCRC has garnered increasing attention. Recent studies indicate that combining traditional Chinese and modern Western medical approaches not only extend survival but also reduces the risk of mortality in patients with BRAF V600E mutant mCRC. This approach is particularly effective for colorectal cancer patients who have right-sided colon involvement, liver metastasis, or a history of radiotherapy or chemotherapy. In this treatment combination, traditional Chinese medicine may offer symptomatic relief and improve quality of life, while Western medicine targets the disease more aggressively with advanced pharmacological agents. Ongoing research is crucial to further elucidate the mechanisms underlying these benefits and to optimize treatment protocols.
Collapse
Affiliation(s)
- Wen-Ming Liu
- Department of Gastrointestinal Surgery, The First People’s Hospital of Tianmen, Tianmen 431700, Hubei Province, China
| | - Xiao-Bing Li
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, Hubei Province, China
| |
Collapse
|
11
|
Jiang C, Chen Z, Yang R, Luogu Z, Ren Q, Hu H, Wang K, Li S, Deng C, Li M, Zheng L. Carbon-Based Flexible Electrode for Efficient Electrochemical Generation of Reactive Chlorine Species in Tumor Therapy. Adv Healthc Mater 2025:e2500369. [PMID: 40411849 DOI: 10.1002/adhm.202500369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/02/2025] [Indexed: 05/26/2025]
Abstract
Reactive chlorine species (RCS) are alternatives to reactive oxygen species (ROS) in tumor therapeutics. Unlike ROS, whose generation is limited by hypoxic conditions or insufficient H2O2 levels in the tumor, RCS can be generated through the electrochemical oxidation of abundant Cl- present in body fluids. However, traditional electrochemical therapy modalities have shown suboptimal outcomes. Herein, a flexible anodic electrode is fabricated by growing a carbon nanowire network (C-NWN) onto carbon cloth (CC). Attributing to its excellent hydrophilicity, high specific surface area, and electrochemical surface area, CC@C-NWN demonstrates a superior capability for RCS generation. Additionally, the carbon vacancies in CC@C-NWN not only enhance Cl- adsorption but also reduce the reaction free energy of the chlorine evolution reaction (CER) more significantly compared to that of the oxygen evolution reaction, thereby promoting the CER process. RCS generated from the CC@C-NWN electrochemical system induces severe oxidative stress, disrupting the redox homeostasis in tumor cells and promoting the synergistic anti-tumor effect of apoptosis and ferroptosis. The pliability of CC@C-NWN enables it to conform closely to the tumor, and it has demonstrated remarkable tumor-suppressive efficacy under low-voltage (3 V) condition in in vivo experiments. Therefore, the work holds significant promise for the development of novel tumor treatment strategies.
Collapse
Affiliation(s)
- Cuinan Jiang
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhaoyu Chen
- National Innovation Center for Industry-Education Integration of Energy Storage Technology, MOE Key Laboratory of Low-Grade Energy Utilization Technologies and Systems, CQU-NUS Renewable Energy Materials & Devices Joint Laboratory, School of Energy and Power Engineering, Chongqing University, Chongqing, 400044, China
| | - Ruihao Yang
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ziga Luogu
- National Innovation Center for Industry-Education Integration of Energy Storage Technology, MOE Key Laboratory of Low-Grade Energy Utilization Technologies and Systems, CQU-NUS Renewable Energy Materials & Devices Joint Laboratory, School of Energy and Power Engineering, Chongqing University, Chongqing, 400044, China
| | - Qian Ren
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hao Hu
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Kaixin Wang
- National Innovation Center for Industry-Education Integration of Energy Storage Technology, MOE Key Laboratory of Low-Grade Energy Utilization Technologies and Systems, CQU-NUS Renewable Energy Materials & Devices Joint Laboratory, School of Energy and Power Engineering, Chongqing University, Chongqing, 400044, China
| | - Senlin Li
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Changlin Deng
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Meng Li
- National Innovation Center for Industry-Education Integration of Energy Storage Technology, MOE Key Laboratory of Low-Grade Energy Utilization Technologies and Systems, CQU-NUS Renewable Energy Materials & Devices Joint Laboratory, School of Energy and Power Engineering, Chongqing University, Chongqing, 400044, China
| | - Lu Zheng
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
12
|
Zhao H, Zhao D, Li S, Liu Y, Zhao R, Zhu X, Xiong P, Mo Y, Gu H, Liu J. PRAP1 regulates colorectal cancer cell proliferation and ferroptosis through the Nrf2 signaling pathway. Cell Signal 2025:111863. [PMID: 40373840 DOI: 10.1016/j.cellsig.2025.111863] [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: 02/24/2025] [Revised: 04/22/2025] [Accepted: 05/11/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a common type of cancer that impacts the digestive tract, and current treatment options have limitations. Studies have confirmed that ferroptosis plays a key role in CRC progression. This research sought to clarify how Proline-rich acidic protein 1 (PRAP1) influences CRC advancement and ferroptosis, and to uncover the underlying mechanisms involved. METHODS Real-time quantitative PCR (RT-qPCR) and western blot were employed to ascertain the levels of PRAP1 in CRC cells (SW480, SW620, and LOVO) and tissues. Immunofluorescence was utilized to locate PRAP1. Biological characterization of CRC cells was determined through CCK-8 assay, EdU staining, Transwell assay, TUNEL staining and Scratch-wound assay. Iron and Fe2+ content was measured using prussian blue staining and iron assay kit. A nude mouse model of xenograft was established, and the impact of PRAP1 on tumor growth was investigated by pathological staining. Expression of ferroptosis-related proteins as well as nuclear factor-erythroid factor 2-related factor 2 (Nrf2) pathway proteins was detected by Western blot. RESULTS PRAP1 levels were elevated in CRC. Overexpression PRAP1 promoted cell proliferation, inhibited apoptosis and ferroptosis. Additionally, overexpression PRAP1 can activate the Nrf2 pathway. However, silencing PRAP1 had the opposite effect. In vivo tumor xenograft experiments showed that silencing PRAP1 resulted in decreased Ki67 positivity and increased TUNEL positivity in tumor tissues, and blocked Nrf2 pathway, thereby inhibited tumor growth. CONCLUSION PRAP1 promotes CRC cell proliferation and inhibits ferroptosis by Nrf2 pathway. This study provides a conceptual framework for the development of novel targeted drugs.
Collapse
Affiliation(s)
- Hongchao Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Deyao Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Siting Li
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Yang Liu
- Endocrinology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China
| | - Ruiwen Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Xiaorong Zhu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Pingping Xiong
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan Province 471003, China
| | - Yingyi Mo
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Hao Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China.
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China.
| |
Collapse
|
13
|
Fu X, Jing Y, Yang R, Zhu C, Tu Y, Hu Z, Sheng W, Jiao Q, Liu L, Zhang W, He Q. Guhan Yangsheng Jing mitigates oxidative stress and ferroptosis to improve reproductive damage in Diabetic Male Rats. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119746. [PMID: 40187624 DOI: 10.1016/j.jep.2025.119746] [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: 02/17/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/07/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE According to traditional Chinese medicine (TCM) theory, reproductive injury is primarily associated with kidney essence deficiency, with the kidney being the affected organ. Guhan Yangshengjing (GHYSJ) is a traditional Chinese patent medicine, its main ingredients of GHYSJ, such as Polygonatum sibiricum Redouté, Epimedium brevicornu Maxim., and Lycium barbarum L. are believed to have significant kidney-tonifying effects, which can improve reproductive damage. AIM OF THE STUDY This study aims to investigate the protective effects of GHYSJ, a traditional Chinese medicine formula, on diabetes-induced male reproductive damage. METHODS In this study, we employed LC/Q-TOF-MS to analyze the active components of GHYSJ. A diabetic rat model was established using a high-sugar high-fat (HSHF) diet in combination with streptozotocin (STZ). Sperm quality and motility were assessed, and testicular morphology and sex hormones (testosterone [T], follicle-stimulating hormone [FSH], and luteinizing hormone [LH]) levels were examined to evaluate the impact of diabetes on reproductive function. Transcriptomic analysis was conducted to elucidate the potential mechanisms underlying GHYSJ's protective effects against diabetes-induced reproductive damage. Additionally, we used ELISA, immunofluorescence, transmission electron microscopy (TEM), immunohistochemistry, and Western blot to measure the expression levels of oxidative stress and ferroptosis-related markers, including oxygen species (ROS), superoxide dismutase (SOD), malondialdehyde (MDA), glutathione (GSH), lipid peroxidation (LPO), ferrous ion (Fe2+), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), glutathione peroxidase 4 (GPX4), and cystine/glutamate antiporter (xCT). RESULTS Diabetic rats induced by a HSHF diet combined with STZ exhibited decreased sperm count, reduced sperm motility, and disrupted sex hormone secretion. GHYSJ intervention significantly reduced ROS levels and MDA accumulation in testicular tissue while enhancing SOD activity, thereby effectively alleviating oxidative damage. Additionally, GHYSJ modulated the Nrf2/HO-1 signaling pathway associated with oxidative stress, restoring testicular antioxidant capacity. This was evidenced by increased GSH levels, upregulated expression of antioxidant proteins (GPX4, xCT), decreased Fe2+ content, and reduced LPO levels. These effects collectively inhibited ferroptosis in testicular tissue of diabetic rats, leading to improved reproductive function. CONCLUSIONS Our findings demonstrate that GHYSJ exerts significant protective effects against diabetes-induced male reproductive damage by modulating oxidative stress and ferroptosis pathways. GHYSJ's ability to enhance antioxidant defenses and inhibit ferroptosis highlights its potential as a therapeutic agent for managing reproductive dysfunction in diabetic males.
Collapse
Affiliation(s)
- Xinying Fu
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China; College of Chinese Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China
| | - Yulan Jing
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China; College of Chinese Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China
| | - Renyi Yang
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China
| | - Congxu Zhu
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China
| | - Yingcen Tu
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China; College of Chinese Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China
| | - Zongren Hu
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China; College of Chinese Medicine, Hunan University of Medicine, Hunan, 418000, China; College of Chinese Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China
| | - Wen Sheng
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China; College of Chinese Medicine, Hunan University of Medicine, Hunan, 418000, China; College of Chinese Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China
| | - Qisen Jiao
- Qidi Pharmaceutical Group Co., Ltd, Hunan, 421099, China
| | - Lumei Liu
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China.
| | - Wei Zhang
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China.
| | - Qinghu He
- College of Integrated Chinese and Western Medicine, Hunan Provincial Key Laboratory for Prevention and Treatment of Heart and Brain Diseases with Integrated Traditional Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China; College of Chinese Medicine, Hunan University of Medicine, Hunan, 418000, China; College of Chinese Medicine, Hunan University of Traditional Chinese Medicine, Hunan, 410208, China.
| |
Collapse
|
14
|
Zhang L, Wang H, Liang B, Qin L, Zhang M, Lv X, Hu S, Fan X, Xie W, Yang H, Huang G, Jing W, Zhao J. Ponicidin promotes ferroptosis to enhance treatment sensitivity in Lenvatinib-resistant hepatocellular carcinoma cells through regulation of KEAP1/NRF2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156824. [PMID: 40382941 DOI: 10.1016/j.phymed.2025.156824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/17/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE This study explores the therapeutic potential of Ponicidin on Lenvatinib-resistant hepatocellular carcinoma (HCC), elucidates its mechanism in reversing Lenvatinib resistance, and provides experimental evidence for its clinical application in overcoming this resistance. METHODS Huh7 and HCC-LM3 cells were used to construct Lenvatinib-resistant cell lines, Huh7-LR and HCC-LM3-LR. Changes in the ferroptosis pathway post-drug resistance were observed by measuring ferroptosis-related markers. The proliferation assay were assessed by CCK-8, while the migration and invasion were measured by scratch and Transwell invasion assays. In mechanistic study, chip analysis and immunoprecipitation with biotin-labeled Ponicidin, were conducted to explore how Ponicidin overcame drug resistance. Xenograft model in nude mice was established to examine Ponicidin's anti-HCC effects In vivo. Clinical specimens were used to assess the true status of patients in Lenvatinib-resistant HCC patients. RESULTS Our study reveals for the first time that ferroptosis inhibition drives Lenvatinib resistance in HCC and identifies Ponicidin as a novel KEAP1-targeting agent to reverse this process. In vitro, ferroptosis pathway was suppressed in Lenvatinib-resistant cells. Ponicidin suppressed proliferation, clonogenicity, migration, and invasion in these cells. The combination of Ponicidin and Lenvatinib significantly inhibited proliferation and reversed drug resistance by activating the ferroptosis pathway. Preliminary mechanistic studies showed that Ponicidin binds to KEAP1, stabilizing the KEAP1/NRF2 interaction, inhibiting the nuclear translocation and activation of NRF2, and thereby inducing ferroptosis to overcome Lenvatinib resistance. In vivo, the combination of Ponicidin and Lenvatinib exhibited a synergistic effect, significantly delaying tumor growth. Clinically, p-NRF2 and GPX4 expression was higher in the Lenvatinib-insensitive group, suggesting that the ferroptosis pathway was inhibited in these patients. Thus, this study demonstrated that Ponicidin promotes ferroptosis to enhances treatment sensitivity in Lenvatinib-resistant HCC cells through KEAP1/NRF2.
Collapse
Affiliation(s)
- Lisha Zhang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China; Nanchang Hongdu Hospital of TCM, Nanchang, PR China
| | - Hao Wang
- Department of Oncology, The Air Force Hosptital of Northern Theater PLA, Shenyang, China
| | - Beibei Liang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Lijuan Qin
- Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Mingzhu Zhang
- University of Shanghai for Science and Technology, Shanghai, PR China
| | - Xingxian Lv
- Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Shi Hu
- Department of Biomedical Engineering, College of Basic Medical Sciences, Second Military Medical University, Shanghai, PR China
| | - Xiaoyu Fan
- Department of Molecular Biology, Shanghai Center for Clinical Laboratory, Shanghai, PR China
| | - Wei Xie
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Wei Jing
- Department of Surgery, Changhai Hospital, Second Military Medical University, Shanghai, PR China.
| | - Jian Zhao
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China.
| |
Collapse
|
15
|
Fan Y, Ma L, Xu X, Fang X, Mauck J, Loor JJ, Sun X, Jia H, Xu C, Xu Q. Heat shock protein B1-mediated ferroptosis regulates mitochondrial dysfunction in adipose tissue of ketotic dairy cows. J Dairy Sci 2025:S0022-0302(25)00300-5. [PMID: 40348370 DOI: 10.3168/jds.2025-26265] [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: 01/03/2025] [Accepted: 04/08/2025] [Indexed: 05/14/2025]
Abstract
In the peripartal period, dairy cow adipose tissue undergo significant metabolic challenges, including oxidative stress and endoplasmic reticulum stress, which could be alleviated by inhibition of ferroptosis. Oxidative stress is often accompanied by mitochondrial damage. However, whether mitochondrial dysfunction occurs in the adipose tissue of ketotic cows are still unclear. Heat shock protein B1 (HSPB1), a key regulator of cellular redox homeostasis, is critical in managing oxidative stress and iron metabolism. Thus, this study aimed to investigate the role of HSPB1-mediated ferroptosis on mitochondrial dysfunction of adipocytes of ketotic dairy cows. We collected adipose tissue samples of clinical ketosis cows (n = 15) with a serum BHB concentration of 3.14 mM (interquartile range = 0.11) and healthy cows (n = 15) with a serum BHB concentration of 0.55 mM (interquartile range = 0.12). Compared with the healthy control group, the protein abundance of HSPB1, transferrin (TF), transferrin receptor 1 (TFR1), 6-transmembrane epithelial antigen of the prostate family member 3 (STEAP3), divalent metal transporter 1 (DMT1), and acyl-CoA synthetase 4 (ACSL4), as well as levels of reactive oxygen species, Fe2+, and total iron were greater in adipose tissue of ketotic cows. Ketotic cows exhibited lower ferroportin (FPN), solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), mitochondrial oxidative phosphorylation complexes I-V (CO I-V), peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), mitofusin-2 (MFN2), nuclear respiratory factor 1 (NRF-1), and mitochondrial transcription factor A (Tfam) protein expression levels, along with lower ATP content compared with control cows. Epinephrine (EPI) treatment upregulated protein abundance of HSPB1 and induced ferroptosis and mitochondrial dysfunction in adipocytes. Inhibition of ferroptosis by pretreatment with ferrostatin-1 (Fer-1) attenuated the EPI-induced decrease in ATP content. Knockdown of HSPB1 by small interfering RNA (si-RNA) exacerbated the EPI-induced upregulation of TF, TFR1, STEAP3, and DMT1 expression and the downregulation of FPN protein expression levels. Furthermore, in the presence of EPI and HSPB1 si-RNA, Fer-1 abolished the regulatory role of HSPB1 on mitochondrial dysfunction, confirming that HSPB1 regulates bovine adipocyte mitochondrial dysfunction in a ferroptosis-dependent manner. Collectively, these data suggest that HSPB1-mediated ferroptosis is an important regulatory mechanism for mitochondrial dysfunction in adipocytes of peripartal dairy cows under negative energy balance.
Collapse
Affiliation(s)
- Yunhui Fan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Li Ma
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Xinyi Xu
- College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514
| | - Xinxin Fang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - John Mauck
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Juan J Loor
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Xudong Sun
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Hongdou Jia
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Chuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Qiushi Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China.
| |
Collapse
|
16
|
Wei X, Fan X, Chai W, Xiao J, Zhao J, He A, Tang X, Li F, Guo S. Dietary limonin ameliorates heart failure with preserved ejection fraction by targeting ferroptosis via modulation of the Nrf2/SLC7A11/GPX4 axis: an integrated transcriptomics and metabolomics analysis. Food Funct 2025; 16:3553-3574. [PMID: 40230319 DOI: 10.1039/d5fo00475f] [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: 04/16/2025]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome characterized by hypertension, metabolic disorders, and impaired diastolic function, with limited therapeutic options. Recent studies have highlighted the role of ferroptosis in the pathogenesis of HFpEF, and the inhibition of ferroptosis occurrence can significantly improve cardiac function. Limonin, a bioactive ingredient derived from citrus fruits, has been confirmed to exert potential anti-inflammatory and antioxidant effects in some cardiovascular diseases. This study aims to investigate the therapeutic effects of limonin on HFpEF and the underlying mechanisms of inhibiting ferroptosis. HFpEF mice were established by a combination of Nω-nitro-L-arginine methyl ester and a high-fat diet for 6 weeks. Subsequently, the HFpEF mice were treated with empagliflozin or limonin via oral gavage for an additional 6 weeks. Limonin curbed body weight gain and improved metabolic disorders and hypertension. Limonin also ameliorated concentric cardiac hypertrophy and diastolic dysfunction. Transcriptomics and metabolomics analyses revealed that limonin regulated ferroptosis-related pathways and lipid peroxidation. In vivo, limonin improved mitochondrial morphology, reduced cardiac Fe2+ levels and ferroptosis markers such as ROS, 4-HNE and MDA, and increased GSH levels, thereby enhancing antioxidant capacity. Mechanistically, limonin regulated the P53/SLC7A11/GPX4 signaling pathway, promoted the nuclear translocation of Nrf2 (its upstream signaling molecule), and subsequently activated its downstream antioxidant elements, ultimately inhibiting ferroptosis. Furthermore, limonin decreased the expressions of ACSL4, COX2, and ALOXs, which reduced the accumulation of lipid peroxides. These results demonstrate that limonin ameliorates HFpEF by targeting ferroptosis via modulation of the Nrf2/SLC7A11/GPX4 axis, providing a novel strategy for HFpEF treatment.
Collapse
Affiliation(s)
- Xiaoqi Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xinyi Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Wangjing Chai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jinling Xiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Jiong Zhao
- Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen 518116, China.
| | - Aolong He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xianwen Tang
- Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen 518116, China.
| | - Fanghe Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Shuzhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
17
|
Shuxia Z, Ping Z, Xiaoyan Z, Sichao M, Xinyi X, Waldron K, Chenfeng W, Rouby SR, Ghonaim AH, Xingxiang C. FB1 causes barrier damage to vascular endothelial cells through ferroptosis by a PINK1/Parkin mediated mitophagy-dependent mechanism. Chem Biol Interact 2025; 416:111536. [PMID: 40324642 DOI: 10.1016/j.cbi.2025.111536] [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: 02/16/2025] [Revised: 04/19/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Fumonisin B1 (FB1) is an environmental mycotoxin produced mainly by fungi of the genus Fusarium. Exposure to FB1 can lead to pulmonary edema in pigs, likely caused by damage to vascular endothelial cells, but the mechanism of FB1-induced damage was unknown. Here, we found that FB1 damages vascular endothelial cells through ferroptosis, marked by iron-dependent membrane lipid peroxidation, and through mitophagy, a selective autophagy that targets mitochondria. FB1 exposure reduced barrier-related gene expression and increased pro-inflammatory factors. Ferroptosis was evidenced by elevated iron, ROS, lipid peroxidation, and ferroptotic markers (TFR, ACSL4), alongside decreased GSH, SLC7A11, and GPX-4 levels in vascular endothelial cells. Importantly, the ferroptosis inhibitor, Ferrostatin-1, reversed the vascular endothelial cells' barrier damage, inflammation, and ferroptosis caused by FB1. FB1-induced mitophagy was demonstrated by detecting decreased mitochondrial membrane potential and increased levels of mitophagy-related proteins. Surprisingly, silencing PINK1 using siRNA not only diminished mitophagy, cellular damage, and inflammatory responses induced by FB1, but also mitigated FB1-induced ferroptosis. In conclusion, this study demonstrates that FB1 causes vascular endothelial cell damage by ferroptosis in a mitophagy-dependent manner. This study thus lays a mechanistic foundation for the study of FB1 causing pulmonary edema in pigs and for exploring options for therapeutic intervention in conditions caused by this mycotoxin, which causes substantial harm to both human and animal health.
Collapse
Affiliation(s)
- Zhang Shuxia
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Zhang Ping
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Zheng Xiaoyan
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Mao Sichao
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Xu Xinyi
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Kevin Waldron
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Wang Chenfeng
- College of Animal Medicine, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Sherin R Rouby
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Ahmed H Ghonaim
- Department of Animal and Poultry Health, Desert Research Center, Cairo, 11435, Egypt
| | - Chen Xingxiang
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
18
|
Pan Y, Xia M, Luo J, Lu S. Resveratrol Promotes Wound Healing by Enhancing Angiogenesis via Inhibition of Ferroptosis. Food Sci Nutr 2025; 13:e70254. [PMID: 40330211 PMCID: PMC12053223 DOI: 10.1002/fsn3.70254] [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: 12/18/2024] [Revised: 04/04/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Diabetic wound healing critically depends on functional endothelial cells for angiogenesis, yet the hyperglycemic microenvironment induces endothelial dysfunction through oxidative stress, inflammation, and senescence. Although ferroptosis has been recognized as a critical pathological factor contributing to impaired diabetic wound healing, the therapeutic potential of resveratrol (Res), a natural polyphenol with well-documented antioxidant and anti-ferroptotic properties, remains underexplored in this context. This study aimed to investigate the protective effects of Res on endothelial cells and elucidate its underlying mechanisms in diabetic wound healing. In vitro experiments systematically evaluated Res's impact on cellular inflammatory responses, senescence levels, and angiogenic capacity. Subsequent in vivo studies assessed Res's therapeutic potential by monitoring diabetic wound healing progression and analyzing associated histological changes. To clarify the mechanisms underlying Res's promotion of diabetic wound healing, we conducted comprehensive analyses measuring intracellular reactive oxygen species, lipid peroxidation levels, mitochondrial membrane potential and morphology, ferroptosis-related marker expression, and upstream signaling pathway regulation. Res significantly reduced HG-induced inflammatory responses and cellular senescence in human umbilical vein endothelial cells while enhancing their angiogenic potential in vitro. In vivo results showed that Res not only markedly accelerated diabetic wound healing but also demonstrated multiple beneficial effects, including effective suppression of cellular senescence, decreased ferroptosis levels, and significantly promoted angiogenesis. Mechanistic investigations confirmed that Res achieves these effects by inhibiting ferroptosis through activation of the PI3K-AKT-Nrf2 signaling axis. Our results demonstrate that Res protects endothelial cells from HG-induced ferroptosis by activating PI3K-AKT-Nrf2 signaling, thereby promoting angiogenesis and diabetic wound healing. These findings highlight Res as a promising therapeutic candidate for impaired diabetic wound repair and justify further clinical investigation.
Collapse
Affiliation(s)
- Yujie Pan
- Department of Traumatic OrthopedicsThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
- School of Clinical Medicine, Guizhou Medical UniversityGuiyangGuizhouChina
| | - Mingyan Xia
- Department of AnatomySchool of Basic Medicine Science, Guizhou Medical UniversityGuiyangChina
| | - Jin Luo
- Department of Traumatic OrthopedicsThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Shuai Lu
- Department of BiologySchool of Basic Medical Science, Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
19
|
Lu Q, Shao N, Fang Z, Ouyang Z, Shen Y, Yang R, Liu H, Cai B, Wei T. The anti-Alzheimer's disease effects of ganoderic acid A by inhibiting ferroptosis-lipid peroxidation via activation of the NRF2/SLC7A11/GPX4 signaling pathway. Chem Biol Interact 2025; 412:111459. [PMID: 40054827 DOI: 10.1016/j.cbi.2025.111459] [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: 01/03/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system, characterized by a gradual decline in cognitive and memory abilities, social disorders, and behavioral abnormalities. Ferroptosis, an iron-dependent type of programmed cell death, is closely associated with the pathogenesis of AD. Ferroptosis is characterized by the accumulation of iron within cells, leading to increased oxidative stress, and ultimately lipid peroxidation and cell death. Ganoderic acid A (GAA), one of the major pharmacologically active components in Ganoderma lucidum, exhibits an excellent neuroprotective effect against AD. However, it is unclear whether GAA improves the symptoms of AD by inhibiting ferroptosis. This study investigated the anti-AD effects of GAA through both in vivo and in vitro experiments, and determined its molecular mechanism from the perspective of ferroptosis modulation. The results showed that GAA administration attenuated hippocampal neuronal loss, improved mitochondrial ultrastructure, and enhanced the memory and learning ability of the AD mice. In vitro assays suggested that GAA effectively protected HT22 AD cells against ferroptosis-related morphological damage, enhanced their antioxidant capacity, maintained their iron metabolism, and reduced mitochondrial dysfunction. Moreover, the immunofluorescence and western blotting results showed that the levels of NFE2 like bZIP transcription factor 2 (NRF2), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11) both in the hippocampus of APP/PS1 mice and amyloid beta (Aβ)25-35-induced HT22 AD cells were markedly enhanced after GAA administration. In summary, these results revealed that GAA improves AD by activating on the NRF2/SLC7A11/GPX4 axis to inhibit ferroptosis-lipid peroxidation.
Collapse
Affiliation(s)
- Qingyang Lu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Nan Shao
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ziyi Fang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Zhaorong Ouyang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yiran Shen
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ruiling Yang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Houli Liu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Biao Cai
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Tao Wei
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
20
|
Ran P, Jiang F, Pan L, Shu Y, Hu F, Wang Y, Zhao R, Wang W, Mu H, Wang J, Wei J, Fu G. Polysaccharide from Atractylodes macrocephala Koidz. alleviates pyrotinib-induced diarrhea through regulating cAMP/LKB1/AMPK/CFTR pathway and restoring gut microbiota and metabolites. Int J Biol Macromol 2025; 308:142512. [PMID: 40157659 DOI: 10.1016/j.ijbiomac.2025.142512] [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: 10/21/2024] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Polysaccharide from Atractylodes macrocephala Koidz. (PAMK), a bioactive component of Atractylodes macrocephala Koidz. (AMK), demonstrates a wide range of pharmacological activities, including the enhancement of gastrointestinal function and regulation of internal homeostasis. This study explores the potential of PAMK in alleviating pyrotinib-induced diarrhea and modulating gut microbiota and its metabolites. Pyrotinib is a tyrosine kinase inhibitor used in cancer treatment, is known for its side effect of diarrhea, which significantly diminishes patients' quality of life. Our prior research suggests that pyrotinib-induced diarrhea may be linked to CFTR-mediated dysregulation of chloride secretion. The present findings indicate that PAMK alleviates pyrotinib-induced diarrhea by reducing cAMP levels, activating the LKB1/AMPK pathway, and inhibiting CFTR activity, as confirmed by enzyme-linked immunosorbent assay (ELISA), qRT-PCR, and western blot analyses. PAMK effectively decreased CFTR-mediated chloride ion secretion in pyrotinib-treated cells, as shown by the MQAE assay. At specific doses, PAMK alleviated pyrotinib-induced diarrhea in rats and significantly restored intestinal barrier integrity. Furthermore, PAMK treatment rebalanced the gut microbiota, reversing the pyrotinib-induced increase in Clostridium and Erysipelotrichi species. Metabolomic profiling further highlighted the involvement of the AMPK signaling pathway. These findings provide a basis for future research aimed at developing cancer treatments with reduced side effects.
Collapse
Affiliation(s)
- Pancen Ran
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China
| | - Fengxian Jiang
- Department of Radiation Oncology, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan 250031, China
| | - Liying Pan
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China
| | - Yang Shu
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China
| | - Fangyan Hu
- School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yahui Wang
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China
| | - Rui Zhao
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Weihao Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Huaiqian Mu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Juqiong Wang
- School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Jian Wei
- School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China.
| | - Guobin Fu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China; Department of Medical Oncology, The Third Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250031, China.
| |
Collapse
|
21
|
Tang J, Zhang J, Yang R, Chen H, Yu X, Peng W, Zeng P. The causal relationships between mitochondria and six types of cancer: a Mendelian randomization study. BMC Cancer 2025; 25:794. [PMID: 40295943 PMCID: PMC12039071 DOI: 10.1186/s12885-025-14201-0] [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: 02/03/2024] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Mitochondria play a multifaceted role in tumorigenesis, influencing energy metabolism, redox balance, and apoptosis. However, whether mitochondrial traits causally affect cancer risk remains unclear. This study aimed to evaluate the potential causal effects of 82 mitochondrial-related exposures on six major cancers-hepatic, colorectal, lung, esophageal, thyroid, and breast-using Mendelian randomization (MR). METHODS Two-sample MR analysis was performed using the inverse variance weighted (IVW) method, with MR-Egger regression and weighted median as complementary approaches. Sensitivity analyses (Cochran's Q test, MR-Egger intercept, leave-one-out) and the Steiger test were applied to assess heterogeneity, pleiotropy, and causal directionality. RESULTS We observed a negative correlation between "39S ribosomal protein L34, mitochondrial", and others, with hepatic cancer, while "[Pyruvate dehydrogenase (acetyl-transferring)] kinase isozyme 2, mitochondrial", and others exhibited a positive correlation with hepatic cancer. "Phenylalanine-tRNA ligase, mitochondrial", and others demonstrated a negative association with colorectal cancer, whereas "Methylmalonyl-CoA epimerase, mitochondrial", and others exhibited a positive correlation with colorectal cancer. "Succinate dehydrogenase assembly factor 2, mitochondrial" exhibited a negative correlation with lung cancer, while "Superoxide dismutase [Mn], mitochondrial levels" showed a positive correlation with lung cancer. "Lon protease homolog, mitochondrial" demonstrated a positive correlation with esophageal cancer. "Iron-sulfur cluster assembly enzyme ISCU, mitochondrial", and others exhibited a negative correlation with thyroid cancer, while "Diablo homolog, mitochondrial", and others showed a positive correlation with thyroid cancer. "ADP-ribose pyrophosphatase, mitochondrial", and others exhibited a negative correlation with breast cancer, while "39S ribosomal protein L34, mitochondrial", and others showed a positive correlation with breast cancer. CONCLUSIONS This study provides MR-based evidence that specific mitochondrial-related traits have causal effects on the risk of several common cancers. Notably, certain single-nucleotide polymorphisms (SNPs) acted as instrumental variables across multiple cancer types through shared mitochondrial mechanisms, such as oxidative stress regulation and metabolic reprogramming. These findings highlight mitochondria as cross-cutting contributors to cancer susceptibility and suggest potential avenues for mitochondrial-targeted prevention and therapy. The identification of pleiotropic genetic variants also offers insights for developing shared biomarkers and therapeutic targets across malignancies.
Collapse
Affiliation(s)
- Jincheng Tang
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jingting Zhang
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Renyi Yang
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Hongyao Chen
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiaopeng Yu
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Wei Peng
- Cancer Research Institute of Hunan Academy of Traditional Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410013, China.
| | - Puhua Zeng
- Cancer Research Institute of Hunan Academy of Traditional Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410013, China.
| |
Collapse
|
22
|
Liu Y, Yang J, Yu F, Li L, Zhao N, Lu C, Lu A, He X. Research advances in traditional Chinese medicine formulae and active components targeting lipid metabolism for hepatocellular carcinoma therapy. Front Pharmacol 2025; 16:1528671. [PMID: 40351413 PMCID: PMC12062747 DOI: 10.3389/fphar.2025.1528671] [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: 11/15/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) has a relatively poor prognosis and a high degree of malignancy. However, the therapeutic drugs are limited. In recent years, abnormal lipid metabolism and its important role in HCC has been reported, and emerging studies found that some formulae and active components of traditional Chinese medicine (TCM) can regulate abnormal lipid metabolism in HCC, showing their good application prospects. Therefore, this article summarizes the changes and the roles of lipid metabolites in HCC progression, and discusses the role of formulae and active components of TCM for the treatment of HCC based on their regulation on abnormal lipid metabolism. A deeper understanding of their relationship may help the precise use of these formulae and active components in HCC.
Collapse
Affiliation(s)
- Yang Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Yang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fenghua Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Shanghai GuangHua Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Saeed BI, Uthirapathy S, Kubaev A, Ganesan S, Shankhyan A, Gupta S, Joshi KK, Kariem M, Jasim AS, Ahmed JK. Ferroptosis as a key player in the pathogenesis and intervention therapy in liver injury: focusing on drug-induced hepatotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04115-w. [PMID: 40244448 DOI: 10.1007/s00210-025-04115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Globally, drug-induced hepatotoxicity or drug-induced liver injury (DILI) is a serious clinical concern. Knowing the processes and patterns of cell death is essential for finding new therapeutic targets since there are not many alternatives to therapy for severe liver lesions. Excessive lipid peroxidation is a hallmark of ferroptosis, an iron-reliant non-apoptotic cell death linked to various liver pathologies. When iron is pathogenic, concomitant inflammation may exacerbate iron-mediated liver injury, and the hepatocyte necrosis that results is a key element in the fibrogenic response. The idea that dysregulated metabolic pathways and compromised iron homeostasis contribute to the development of liver injury by ferroptosis is being supported by new data. Various ferroptosis-linked genes and pathways have been linked to liver injury, although the molecular processes behind ferroptosis's pathogenicity are not well known. Here, we delve into the features of ferroptosis, the processes governing ferroptosis, and our current knowledge of iron metabolism. We also provide an overview of ferroptosis's involvement in the pathophysiology of liver injury, particularly DILI. Lastly, the therapeutic possibilities of ferroptosis targeting for liver injury management have been provided. Natural products, nanoparticles (NPs), mesenchymal stem cell (MSC), and their exosomes have attracted increasing attention among such therapeutics.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
24
|
Huang R, Xu R, Shi J, Yang Z, Zheng J, Wei D. Artesunate induces ferroptosis in osteosarcoma through NCOA4-mediated ferritinophagy. FASEB J 2025; 39:e70488. [PMID: 40168090 PMCID: PMC11960798 DOI: 10.1096/fj.202403160r] [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: 12/10/2024] [Revised: 02/20/2025] [Accepted: 03/14/2025] [Indexed: 04/03/2025]
Abstract
Osteosarcoma (OS) is a prevalent primary malignant bone tumor that lacks effective therapeutic interventions. Artesunate (ART) has been proved to have remarkable treatment effects on severe malaria and anti-tumor properties. This study aimed to investigate the anti-OS effects and underlying mechanisms of ART. The potential mechanisms of ART-mediated anti-OS activity were analyzed by using RNA sequencing, iron accumulation, lipid peroxidation, western blotting, and small interfering RNA (siRNA) transfection. In vivo, a xenograft mice model was adopted to explore the anticancer effect of ART. The present study revealed that ART significantly suppressed OS cell proliferation. Subsequent results suggested that ART exerted anti-OS activity mainly through the ferroptosis pathway. ART decreased the GSH/GSSG ratio, xCT and GPX4 expression, while increasing MDA and lipid peroxidation, which were reversed by Fer-1, DFO, 3-MA, and NCOA4 silencing. Mechanistically, ART upregulated the expression of TFR and DMT1, and triggered ferritinophagy by upregulating the expression of NCOA4, which increased Fe2+ accumulation and triggered ferroptosis. In addition, cytoplasmic iron further activated Mfrn2-mediated transportation of cytoplasmic free iron into the mitochondria, resulting in mitochondrial iron overload, eventually leading to lipid peroxidation and ferroptosis. Furthermore, in an OS xenograft mouse model, administration of ART inhibited tumor growth by ferroptosis. Collectively, our findings indicated that ART has the potential anti-OS capacity through NCOA4-mediated ferritinophagy, which might shed light on the future of OS therapy.
Collapse
Affiliation(s)
- Rui Huang
- Department of Orthopedic SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuanChina
- The First School of Clinical MedicineGeneral Hospital of Ningxia Medical UniversityYinchuanChina
| | - Ruiqing Xu
- Department of Orthopedic SurgeryHonghui Hospital, Xi'an Jiaotong UniversityXi'anChina
| | - Jiandang Shi
- Department of Orthopedic SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuanChina
| | - Zongqiang Yang
- Department of Orthopedic SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuanChina
- The First School of Clinical MedicineGeneral Hospital of Ningxia Medical UniversityYinchuanChina
| | - Jianping Zheng
- Department of Orthopedic SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuanChina
| | - Daihao Wei
- Department of Orthopedic SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuanChina
- The First School of Clinical MedicineGeneral Hospital of Ningxia Medical UniversityYinchuanChina
| |
Collapse
|
25
|
Du H, Yang K, Yang J, Wan J, Pan Y, Song W, Xu S, Chen C, Li J. Euphorbia humifusa Willd. ex Schltdl. Mitigates Liver Injury via KEAP1-NFE2L2-Mediated Ferroptosis Regulation: Network Pharmacology and Experimental Validation. Vet Sci 2025; 12:350. [PMID: 40284852 PMCID: PMC12030869 DOI: 10.3390/vetsci12040350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Liver injury poses major health risks in livestock, necessitating effective therapeutic interventions. This study elucidates the hepatoprotective mechanisms of Euphorbia humifusa Willd. ex Schltdl. (EHW) by integrating network pharmacology, molecular docking, and experimental validation. Using a CCl4-induced liver injury model mimicking veterinary clinical scenarios, EHW markedly alleviated hepatic damage, demonstrated by reduced liver index, serum ALT and AST levels, histopathological lesions, iron accumulation, inflammatory cytokines, and ferroptosis-associated gene expression. Network pharmacology identified EHW's core bioactive components (quercetin, kaempferol, and β-sitosterol) and critical targets (IL-6, STAT3, HIF-1α, PTGS2, NFE2L2, and KEAP1) which were linked to ferroptosis and oxidative stress. Molecular docking revealed robust binding affinities between these compounds and ferroptosis-related proteins. In vivo validation confirmed that EHW inhibited KEAP1, activated NFE2L2-mediated antioxidant defenses (upregulating SOD1 and NQO1), restored iron homeostasis (lowering TFR1, elevating FTH1), and attenuated phospholipid peroxidation by suppressing ACSL4 and ALOX12. These results indicate that EHW mitigates ferroptosis-driven liver injury via KEAP1-NFE2L2 signaling to restore iron homeostasis and reduce oxidative stress, offering a mechanistic foundation for its clinical application in veterinary hepatoprotection.
Collapse
Affiliation(s)
- Hongxu Du
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
- Institute of Traditional Chinese Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Kunzhao Yang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Jingyi Yang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Junjie Wan
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Yu Pan
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Weijie Song
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Shuang Xu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Cheng Chen
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Jiahui Li
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| |
Collapse
|
26
|
Wang C, Zhao Y, Peng Y, Chen W, Zhu J, Gu C, Huo R, Ding L, Liu Y, Liu T, Zhang C, Yang W, Wang H, Guo W, Wang B. Juglone induces ferroptotic effect on hepatocellular carcinoma and pan-cancer via the FOSL1-HMOX1 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156417. [PMID: 39923427 DOI: 10.1016/j.phymed.2025.156417] [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: 10/15/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Drug therapy plays an essential role in the management of hepatocellular carcinoma (HCC). Recently, the use of natural products to suppress tumor cells has emerged as a promising direction for drug development. Juglone, a natural compound, exhibits anticancer activities across various cancer types. However, the precise mechanism underlying the anticancer effect of juglone, especially in HCC, remains elusive. PURPOSE This study aimed to investigate the potential inhibitory effects of juglone on HCC and pan-cancer, as well as elucidate the underlying mechanism. METHODS Cell Counting Kit-8 and colony formation assays were used to examine cell proliferation. Transwell and wound healing assays were used to evaluate cell migration. Cell cycle distribution was assessed by flow cytometry. The in vivo effect of juglone on HCC was evaluated by establishing the HCC xenograft mice model. RNA sequencing and inhibitors targeting diverse modes of programmed cell death were applied to uncover the form of juglone-induced cell death. Integrated transcriptomic, and proteomic analyses unveiled the underlying mechanism. The dual-luciferase reporter assay was employed to verify the findings. The pan-cancer value of juglone was assessed using TCGA database analysis and cellular assays. RESULTS Juglone suppressed HCC growth via ferroptosis in vitro and in vivo, which is evidenced by increased levels of iron, lipid peroxidation (LPO), reactive oxygen species (ROS), malondialdehyde (MDA), and decreased levels of glutathione (GSH). Omic analyses, gene silencing and functional analyses showed the upregulated HMOX1 and FOSL1 were the key effector molecule and transcriptional factor in juglone-induced ferroptosis, respectively. The binding site of FOSL1 at the promoter of HMOX1 was identified. Juglone could induce ferroptosis in pan-cancer by activating the FOSL1-HMOX1 axis. CONCLUSION Our findings, for the first time, demonstrate that juglone effectively inhibits tumor growth by inducing FOSL1-HMOX1-dependent ferroptosis, thereby offering a promising strategy for the development of anticancer drugs.
Collapse
Affiliation(s)
- Chuyu Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ying Zhao
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yingfei Peng
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Wei Chen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Jie Zhu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Chenzheng Gu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ran Huo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Lin Ding
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yu Liu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Chunyan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China; Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, PR China; Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, PR China; Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, PR China
| | - Wenjing Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Hao Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China; Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, PR China; Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, PR China; Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, PR China.
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China; Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, PR China; Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, PR China; Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, PR China.
| |
Collapse
|
27
|
Zhu Y, Huang F, Liu X, Hou Y, Huang Y. Phillyrin regulates the JAK2/STAT3 signaling pathway by inhibiting TOP2A expression to accelerate ferroptosis in hepatocellular carcinoma. Oncol Rep 2025; 53:43. [PMID: 39950325 PMCID: PMC11843411 DOI: 10.3892/or.2025.8876] [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: 10/18/2024] [Accepted: 01/14/2025] [Indexed: 02/23/2025] Open
Abstract
Despite advancements and refinements in the therapeutic approaches for hepatic malignancies, liver cancer remains a prevalent and deadly form of cancer, with its grim outlook posing as a significant clinical challenge. Phillyrin (PHN) has been reported to have anticancer effects, but the anticancer mechanism in liver cancer is ominous. By searching the potential target of PHN in the online database and liver cancer disease database, it was found that there is only one overlap gene, and DNA topoisomerase II alpha (TOP2A) is abnormally expressed in liver cancer tissues. TOP2A overexpression and downregulated hepatocellular carcinoma cell lines were then constructed in vitro, and it was examined whether PHN treatment induced ferroptosis in hepatocellular carcinoma by regulating TOP2A's inhibition of Janus kinase 2/Signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway through phenotypic assay, western blot assay, reverse transcription‑quantitative PCR assay and electron microscopy. The results showed that PHN could inhibit the expression of TOP2A protein and JAK2/STAT3 signaling pathway in hepatoma cells. PHN could also downregulate glutathione peroxidase 4 by suppressing the expression of TOP2A protein. PHN impeded the activity of factor inhibiting hypoxia‑inducible factor 1 alpha, thereby augmenting the synthesis of iron‑dependent apoptosis‑related proteins including cytochrome c oxidase subunit II, long‑chain acyl‑CoA synthetase family member 4 and NADPH oxidase 1, thus facilitating an increase in Fe2+ concentration and accelerating oxidative harm within hepatocellular carcinoma cells, culminating in the induction of ferroptotic cell death in these liver malignancy cells.
Collapse
Affiliation(s)
- Ying Zhu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Fenghe Huang
- Hebei Yiling Medical Research Institute Co., LTDS, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yunlong Hou
- Hebei Yiling Medical Research Institute Co., LTDS, Shijiazhuang, Hebei 050000, P.R. China
| | - Yong Huang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
28
|
Yuan F, Han S, Li Y, Li S, Li D, Tian Q, Feng R, Shao Y, Liang X, Wang J, Lei H, Li X, Duan Y. miR-214-3p attenuates ferroptosis-induced cellular damage in a mouse model of diabetic retinopathy through the p53/SLC7A11/GPX4 axis. Exp Eye Res 2025; 253:110299. [PMID: 39978746 DOI: 10.1016/j.exer.2025.110299] [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: 12/17/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Ferroptosis has been implicated in the development of diabetic retinopathy (DR). This study aimed to identify novel ferroptosis-related regulators involved in the pathophysiology of DR using an in vivo streptozotocin (STZ)-induced diabetic model in C57BL/6J mice and cultured primary human retinal vascular endothelial cells (HRECs). Transmission electron microscopy revealed mitochondrial morphological changes consistent with ferroptosis in vascular endothelial cells from STZ-treated mice. Western blot analysis showed increased levels of ferroptosis markers (4-HNE, p53, phosphorylated p53) along with decreased levels of glutathione (GSH), SLC7A11, and GPX4 in diabetic mice. In vitro experiments demonstrated that ferroptosis inhibitors, including pifithrin-α (a p53 inhibitor) and ferrostatin-1 (Fer-1), mitigated cellular damage and Fe2+ accumulation in high-glucose-treated HRECs. These inhibitors also improved mitochondrial membrane potential and restored GSH levels. Bioinformatics analysis and dual-luciferase assays identified a p53 binding site within the miR-214-3p sequence. Overexpression of miR-214-3p in high-glucose-treated HRECs resulted in downregulation of p53 and upregulation of SLC7A11 and GPX4, thereby alleviating ferroptosis-induced injury. This study demonstrates that ferroptosis contributes to retinal damage at tissue, cellular, and molecular levels in DR. Specifically, p53, regulated by miR-214-3p, promotes ferroptosis through the SLC7A11/GPX4 pathway under high-glucose conditions. These findings suggest that the miR-214-3p/p53/SLC7A11/GPX4 axis could serve as a potential therapeutic target for managing ferroptosis and retinal damage in diabetic retinopathy.
Collapse
Affiliation(s)
- Fang Yuan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China; Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Songyu Han
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yahong Li
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sanming Li
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Dian Li
- Washington University in St. Louis, MO, USA
| | - Qingjun Tian
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ronghua Feng
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Shao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xing Liang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingbo Wang
- Shanxi Eye Hospital Affiliated with Shanxi Medical University, Taiyuan, 030072, China
| | - Hetian Lei
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.
| | - Yajian Duan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China; Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
29
|
Xiong Z, Sui X, Bai Y, Liu Y, Leng Y, Wang S, Su B, Liu Z, Liu T. Hua Zheng San Ji Fang suppresses liver cancer progression by inhibiting TYRO3 expression via the ERK signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156497. [PMID: 40023065 DOI: 10.1016/j.phymed.2025.156497] [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: 10/18/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Liver cancer poses a significant global health challenge owing to its increasing incidence and associated mortality rates. Traditional Chinese Medicine (TCM) has garnered attention for its potential in oncology, with formulations such as Hua Zheng San Ji Fang (HZSJF) exhibiting antineoplastic effects. HZSJF is clinically employed in China for cancer treatment; however, its molecular mechanisms in liver cancer remain elusive. TYRO3 plays a key role in tumor progression via the ERK signaling pathway, rendering it a potential therapeutic target. However, the effect of HZSJF on TYRO3 expression and its downstream signaling in liver cancer remains unexplored. PURPOSE This study aimed to investigate the molecular mechanisms through which HZSJF alleviates liver cancer progression, focusing on its regulation of TYRO3 and the ERK signaling pathway. METHODS TYRO3 expression in liver cancer and para-carcinoma tissues was analyzed using immunohistochemistry, reverse transcription-quantitative PCR, and western blotting. Liver cancer cells were used to investigate HZSJF-regulated pathways. Transcriptome sequencing was used to identify HZSJF-targeted genes. Cell proliferation, apoptosis, invasion, and migration were assessed using EdU, YO-PRO-1/PI staining, and transwell assays. ERK signaling involvement was examined using a specific inhibitor and validated in vivo using subcutaneous nude mouse tumor models. RESULTS HZSJF significantly inhibited TYRO3 expression and ERK pathway activation, reducing proliferation, invasion, and migration while promoting apoptosis. The ERK inhibitor corroborated the pathway's role in the antitumor effects of HZSJF. HZSJF suppressed tumor growth and TYRO3 expression in vivo. CONCLUSION HZSJF alleviated liver cancer progression through the ERK signaling pathway by inhibiting TYRO3 expression, presenting a potential therapeutic approach.
Collapse
Affiliation(s)
- Zhuang Xiong
- Changchun University of Chinese Medicine, 1035 Bo Shuo Road, Changchun City, Jilin Province, China; Department of Liver, Spleen and Gastroenterology, First Affiliated Hospital to Changchun University of Chinese Medicine, 1478 Gong Nong Road, Changchun City, Jilin Province, China.
| | - Xiaodan Sui
- Changchun University of Chinese Medicine, 1035 Bo Shuo Road, Changchun City, Jilin Province, China; Department of Liver, Spleen and Gastroenterology, First Affiliated Hospital to Changchun University of Chinese Medicine, 1478 Gong Nong Road, Changchun City, Jilin Province, China.
| | - Yu Bai
- Changchun University of Chinese Medicine, 1035 Bo Shuo Road, Changchun City, Jilin Province, China; ICU, Changchun University of Traditional Chinese Medicine Affiliated Third Clinical Hospital, 1643 Jing Yue Street, Changchun City, Jilin Province, China.
| | - Yangyang Liu
- Changchun University of Chinese Medicine, 1035 Bo Shuo Road, Changchun City, Jilin Province, China; Preventive Medicine Department, First Affiliated Hospital to Changchun University of Chinese Medicine, 1478 Gong Nong Road, Changchun City, Jilin Province, China.
| | - Yan Leng
- Changchun University of Chinese Medicine, 1035 Bo Shuo Road, Changchun City, Jilin Province, China; Department of Liver, Spleen and Gastroenterology, First Affiliated Hospital to Changchun University of Chinese Medicine, 1478 Gong Nong Road, Changchun City, Jilin Province, China.
| | - Song Wang
- Changchun University of Chinese Medicine, 1035 Bo Shuo Road, Changchun City, Jilin Province, China; Department of Liver, Spleen and Gastroenterology, First Affiliated Hospital to Changchun University of Chinese Medicine, 1478 Gong Nong Road, Changchun City, Jilin Province, China.
| | - Boyang Su
- Changchun University of Chinese Medicine, 1035 Bo Shuo Road, Changchun City, Jilin Province, China.
| | - Zhiyuan Liu
- Changchun University of Chinese Medicine, 1035 Bo Shuo Road, Changchun City, Jilin Province, China.
| | - Tiejun Liu
- Changchun University of Chinese Medicine, 1035 Bo Shuo Road, Changchun City, Jilin Province, China; Department of Liver, Spleen and Gastroenterology, First Affiliated Hospital to Changchun University of Chinese Medicine, 1478 Gong Nong Road, Changchun City, Jilin Province, China.
| |
Collapse
|
30
|
Han L, Zhai W. Mechanisms and preventive measures of ALDH2 in ischemia‑reperfusion injury: Ferroptosis as a novel target (Review). Mol Med Rep 2025; 31:105. [PMID: 40017132 PMCID: PMC11876945 DOI: 10.3892/mmr.2025.13470] [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: 10/17/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
Ischemia‑reperfusion injury (IRI) refers to tissue or organ damage that occurs following a period of inadequate blood supply (ischemia) followed by restoration of blood flow (reperfusion) within a short time frame. This phenomenon is prevalent in clinical conditions such as cardiovascular and cerebrovascular disease, organ transplantation and stroke. Despite its frequency, effective therapeutic strategies to mitigate IRI remain elusive in clinical practice, underscoring the need for a deeper understanding of its molecular mechanisms. Aldehyde dehydrogenase 2 (ALDH2), a key enzyme in alcohol metabolism, serves a role in alleviating oxidative stress and cell damage during IRI by modulating oxidative stress, decreasing apoptosis and inhibiting inflammatory responses. ALDH2 exerts protective effects by detoxifying reactive aldehydes, thereby preventing lipid peroxidation and maintaining cellular homeostasis. Furthermore, ferroptosis, a regulated form of cell death driven by iron accumulation and subsequent lipid peroxidation, is a key process in IRI. However, the precise role of ALDH2 in modulating ferroptosis during IRI remains incompletely understood. Although there is an interaction between ALDH2 activity and ferroptosis, the underlying mechanisms have yet to be clarified. The present review examines the role of ALDH2 and ferroptosis in IRI and the potential regulatory influence of ALDH2 on ferroptosis mechanisms, as well as potential targeting of ALDH2 and ferroptosis for IRI treatment and prevention. By elucidating the complex interplay between ALDH2 and ferroptosis, the present review aims to provide new insights for the development of innovative therapeutic strategies to mitigate ischemic tissue damage and improve clinical outcomes.
Collapse
Affiliation(s)
- Liang Han
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, P.R. China
| | - Wen Zhai
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
31
|
Go MY, Kim J, Jeon CY, Kim M, Shin DW. Pinitol Improves Lipopolysaccharide-Induced Cellular Damage in Human Dermal Microvascular Endothelial Cells. Molecules 2025; 30:1513. [PMID: 40286119 PMCID: PMC11990420 DOI: 10.3390/molecules30071513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
3-O-Methyl-D-chiro-inositol (pinitol) has been reported to possess insulin-like effects and is known as one of the anti-diabetic agents for improving muscle and liver function. However, the beneficial effects of pinitol on human dermal microvascular endothelial cells (HDMECs) are not well understood. In this study, we investigated whether pinitol could protect HDMECs from damage induced by lipopolysaccharides (LPSs), which cause various cell defects. We observed that pinitol enhanced wound healing for LPS-damaged HDMECs. We found that pinitol significantly downregulated the LPS-induced upregulation of reactive oxygen species (ROS). Pinitol also significantly restored the mitochondrial membrane potential in these cells. Immunofluorescence analysis revealed that pinitol notably reduced the nuclear localization of NF-κB in LPS-damaged HDMECs. Furthermore, we demonstrated that pinitol decreased the phosphorylation levels of the MAPK family in LPS-damaged HDMECs. Interestingly, we observed that pinitol improved tube formation in LPS-damaged HDMECs. Taken together, we suggest that pinitol exerts several beneficial effects on LPS-damaged HDMECs and may be a promising therapeutic agent for improving vascular-related skin diseases.
Collapse
Affiliation(s)
| | | | | | | | - Dong Wook Shin
- Research Institute for Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea; (M.Y.G.); (J.K.); (C.Y.J.); (M.K.)
| |
Collapse
|
32
|
Gu M, Sun L, Yang J, Wang K, Wu F, Zheng L, Shen X, Lai X, Gong L, Peng Y, Xu S, Yang J, Yang C. Ditan Decoction ameliorates vascular dementia-induced cognitive dysfunction through anti-ferroptosis via the HIF1α pathway: Integrating network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119459. [PMID: 39978449 DOI: 10.1016/j.jep.2025.119459] [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: 11/08/2024] [Revised: 01/06/2025] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Vascular dementia (VaD) represents a frequently seen cognitive dysfunction syndrome and has ranks second among dementia subtypes following Alzheimer's disease. At present, Ditan Decoction (DTD), the traditional Chinese herbal prescription, is clinically applied in treating VaD. However, the material basis of its efficacy and therapeutic mechanism still remain unknown. AIM OF THE STUDY This experiment investigated the protection induced by DTD against VaD and the associated mechanism through network pharmacology, mass spectrometry analysis, and in vivo validation. MATERIALS AND METHODS We induced VaD in a rat model using bilateral common carotid artery ligation method (2-VO) and administered DTD at doses of 2.14, 4.28 and 8.55 g/kg, with Memantine (0.9 mg/kg) being the positive control. Following oral administration with DTD or Memantine for 4 weeks, behavioral tests were used for assessing cognitive function. H&E and Nissl staining was used for evaluating hippocampal pathology. TEM was used to visualize the ultrastructure of the hippocampal tissue. ELISA was carried out for measuring inflammatory factor levels in rat serum, and biochemical assays were employed to assess oxidative stress levels. Ferroptosis in the hippocampus was examined through analyzing corresponding biomarkers and protein expression. Additionally, HPLC-Q-Exactive-MS technology was employed for identifying DTD components, whereas network pharmacology was conducted for predicting DTD's targets for treating VaD. HIF1α expression levels were assessed by Western blotting and immunofluorescence. We also further validated whether the protective effects of DTD on VaD were mediated through the HIF1α-regulated ferroptosis signaling pathway by using an HIF1α inhibitor in rats. RESULTS DTD demonstrated protective effects against 2VO-induced hippocampal injury through alleviating oxidative stress, lowering systemic inflammation, while preventing ferroptosis of hippocampal tissue. As revealed by network pharmacology, DTD probably executes its function in VaD by activating HIF1α pathway. According to immunofluorescence and Western blotting, DTD activated HIF1α within hippocampal tissue. Furthermore, DTD's protection against VaD and ferroptosis was reversed when an HIF1α inhibitor was applied. CONCLUSION These findings suggested that DTD rescued cognitive dysfunction in VaD by inhibiting ferroptosis via activating HIF1α pathway.
Collapse
Affiliation(s)
- Mengyu Gu
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Lieqian Sun
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Jie Yang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Kaiyi Wang
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Fan Wu
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Li Zheng
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Xiangzhong Shen
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Xing Lai
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Lili Gong
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Ying Peng
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Shujie Xu
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Jia Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Chao Yang
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China.
| |
Collapse
|
33
|
Xu M, Trung TS, Zhu Z, Li S, Gong S, Cheng N, Zhou P, Wang S. ESR1-dependent suppression of LCN2 transcription reverses autophagy-linked ferroptosis and enhances sorafenib sensitivity in hepatocellular carcinoma. J Physiol Biochem 2025:10.1007/s13105-025-01073-y. [PMID: 40126852 DOI: 10.1007/s13105-025-01073-y] [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: 10/22/2024] [Accepted: 03/05/2025] [Indexed: 03/26/2025]
Abstract
Sorafenib resistance is a significant hurdle in the treatment landscape of hepatocellular carcinoma (HCC). Lipocalin 2 (LCN2), a secretory glycoprotein that transports lipophilic molecules across cell membranes, is thought to affect the s therapeutic efficacy of sorafenib. Despite its importance, the detailed regulatory pathways involving LCN2 are still being deciphered. We probed the correlation between LCN2 expression and sorafenib resistance in HCC cells. Through the modulation of LCN2 levels, we investigated its role in cell proliferation, apoptosis, and its regulatory effects on autophagy-driven ferroptosis. With the aid of hTFtarget and JASPAR databases, ESR1 was pinpointed as a transcriptional inhibitor of LCN2. The impact of the ESR1-LCN2 axis on sorafenib resistance in HCC was then examined in vitro and validated in a xenograft tumor mouse model. In HCC cells, elevated LCN2 levels were found to be associated with resistance to sorafenib. Depletion of LCN2 resulted in attenuated HCC cell growth and elevated rates of apoptosis and ferroptosis. Overexpression of LCN2 had the opposite effect, promoting cell proliferation and suppressing cell death pathways, a response that could be overridden by autophagy agonists. ESR1 suppressed LCN2 transcription, which in turn activated autophagy-mediated ferroptosis, mitigating sorafenib tolerance in HCC and enhancing the therapeutic index. ESR1 targets LCN2 transcription to initiate autophagy-driven ferroptosis, thereby reducing sorafenib resistance in HCC cells.
Collapse
Affiliation(s)
- Mingfang Xu
- Department of Otolaryngology Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434020, China
| | - Tran Sy Trung
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Zhiyong Zhu
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Shijia Li
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Shicheng Gong
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Nuo Cheng
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Peng Zhou
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Shuai Wang
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China.
| |
Collapse
|
34
|
Liu C, Deng Y, Huang L, Nie X, Jiang Y, Zhang X, Zhang H. USP5 Suppresses Ferroptosis in Bladder Cancer Through Stabilization of GPX4. Curr Issues Mol Biol 2025; 47:211. [PMID: 40136465 PMCID: PMC11941033 DOI: 10.3390/cimb47030211] [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: 02/28/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
USP5 has been proven to play an important role in the proliferation of bladder cancer (BC). In this study, we focused on investigating the molecular mechanism of ferroptosis induced by USP5 in bladder cancer. The role of USP5 in bladder cancer was evaluated using T24 wild-type cells (WT) and USP5 knockout (USP5-/-) by CCK8 and colony formation assays. The contents of ferrobivalent ions (Fe2+), reactive oxygen species (ROS), and malondialdehyde (MDA) were detected using a determination kit to observe the relationship between USP5 and ferroptosis. Furthermore, the molecular mechanism study was evaluated by employing Western blotting, co-immunoprecipitation, RT-qPCR, ubiquitination assays, etc. This study showed genetic ablation of USP5 significantly inhibited the viability and proliferation of bladder cancer cells. Genetic ablation of USP5 promoted increases in Fe2+ content, ROS, and MDA levels. The addition of erastin significantly increased the viability and proliferation of T24 USP5-/- cells and significantly increased their ROS and MDA contents. We verified that USP5 deficiency led to a significant reduction in GPX4 protein levels and that the overexpression of USP5 could stabilize the GPX4 protein. Further studies showed that USP5 interacts with GPX4 and stabilizes GPX4 by inhibiting its ubiquitination These findings revealed USP5 inhibits ferroptosis in bladder cancer cells by stabilizing GPX4. The relationship between USP5 and ferroptosis could be a potential therapeutic target for bladder cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Xia Zhang
- School of Medical Technology and Translational Medicine, Hunan Normal University, 371 Tongzipo Road, Yuelu District, Changsha 410013, China; (C.L.); (Y.D.); (L.H.); (X.N.); (Y.J.)
| | - Huihui Zhang
- School of Medical Technology and Translational Medicine, Hunan Normal University, 371 Tongzipo Road, Yuelu District, Changsha 410013, China; (C.L.); (Y.D.); (L.H.); (X.N.); (Y.J.)
| |
Collapse
|
35
|
Guo L, Zhang J, Li Y, Gao Y, Huang J, Liu M, Li J, Chai W, Li Y. 3,3'-diindolylmethane induces ferroptosis and inhibits proliferation in non-small-cell lung cancer through the AHR/NRF2/GPX4 axis. Discov Oncol 2025; 16:344. [PMID: 40100489 PMCID: PMC11920504 DOI: 10.1007/s12672-025-02096-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/07/2025] [Indexed: 03/20/2025] Open
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor. In lung cancer, AHR activation stimulates cancer cell proliferation and promotes tissue invasion and metastasis, and targeting the AHR pathway is an effective way to prevent and treat lung cancer. In lung cancer, AHR binds to the NRF2 promoter region to promote carcinogenesis, but treatment research based on AHR/NRF2 pathway is insufficient. 3,3'-diindolylmethane (DIM), an active phytochemical derivative extracted from cruciferous vegetables, is a modulator of AHR. In this study, We investigated the medicinal value of DIM in NSCLC (non-small cell lung cancer) by in vivo and in vitro experiments and explored the underlying mechanisms. In vitro studies showed that DIM inhibited the viability of NSCLC and induced apoptosis and cycle arrest in cancer cells. DIM inhibited the migration and invasion of NSCLC cells by reversing the epithelial-mesenchymal transition. DIM induced ferroptosis in NSCLC cells; increased cellular Fe2+, ROS (reactive oxygen species), and MDA; decreased cellular GSH, AHR, NRF2, and GPX4; and disrupted the mitochondrial membrane potential. The effect of DIM-induced ferroptosis can be reversed by the AHR receptor antagonist CH-223191, ferroptosis inhibitor Fer-1, and ROS scavenger NAC. Overexpression of NRF2 reversed DIM-induced ferroptosis. Identical results were obtained in a nude mouse xenograft model. In summary, we have confirmed that DIM has significant potential in the treatment of non-small cell lung cancer. DIM induces cancer cell ferroptosis through the AHR/NRF2/GPX4 axis. These findings provide experimental basis for DIM treatment and future clinical research in non-small cell lung cancer.
Collapse
Affiliation(s)
- Lin Guo
- Department of Respiratory Medicine, the First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5Renmin Street, Guta District, Jinzhou City, 121001, Liaoning Province, China
| | - Jia Zhang
- Department of Hematology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Yuqiang Li
- Clinical Biobank Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Yuehong Gao
- Liaoyang Chest Hospital, Emei Committee, Shuguang Town, Hongwei District, Liaoyang City, Liaoning Province, China
| | - Jiali Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5Renmin Street, Guta District, Jinzhou City, 121001, Liaoning Province, China
| | - Mengru Liu
- Department of Respiratory Medicine, the First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5Renmin Street, Guta District, Jinzhou City, 121001, Liaoning Province, China
| | - Jing Li
- Department of Respiratory Medicine, the First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5Renmin Street, Guta District, Jinzhou City, 121001, Liaoning Province, China
| | - Wenshu Chai
- Department of Respiratory Medicine, the First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5Renmin Street, Guta District, Jinzhou City, 121001, Liaoning Province, China.
| | - Yubin Li
- Key Surgical Laboratory of Educational Administration of Liaoning Province, the First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5Renmin Street, Guta District, Jinzhou City, 121001, Liaoning Province, China.
| |
Collapse
|
36
|
Chen R, Wen L, Guo F, He J, Wong KH, Chen M. Glutathione-scavenging natural-derived ferroptotic nano-amplifiers strengthen tumor therapy through aggravating iron overload and lipid peroxidation. J Control Release 2025; 379:866-878. [PMID: 39842724 DOI: 10.1016/j.jconrel.2025.01.026] [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: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025]
Abstract
Nanomedicine-driven ferroptosis has emerged as a promising tumor treatment strategy through delivering exogenous iron and aggravating the lethal accumulation of lipid peroxides (LPO). However, the compensatory mechanisms of ferroptosis defense systems in cancer cells compromise the therapeutic efficacy and lead to potential side effects. Herein, a highly effective ferroptotic nano-amplifier is designed to synergistically promote ferroptosis via increasing intracellular labile iron, exacerbating lipid peroxidation and overcoming the defense system. Briefly, a natural-derived amphiphilic polymer composing of chondroitin sulfate (CS), arachidonic acid (AA) and a redox-sensitive linker, cystamine (CYS) is constructed to self-assemble as a GSH-responsive nanodrug delivery system for loading bioactive ingredient Polyphyllin I (PPI) and ferric ion (Fe3+). This nanodrug (CSAA/Fe@PPI) can scavenge the aberrant intracellular GSH via CYS linker, accompanied with the degradation of CSAA/Fe@PPI and the release of PPI, AA and Fe3+. On one hand, the intracellular labile iron level is significantly elevated due to the exogenous delivery of Fe3+ and PPI-induced ferritinophagy. On the other hand, ROS burst and the supplement of AA initiate and propagate the lipid peroxidation chain reaction. Meanwhile, the depletion of intracellular GSH suppresses the GPX4 activity, further strengthening the lethal accumulation of LPO. Consequently, the ferroptotic antitumor efficacy is remarkably improved by systemically aggravating iron overload and lipid peroxidation. Therefore, our study presents an effective strategy to improve ferroptosis-based anti-cancer treatment through multiple intervention routes.
Collapse
Affiliation(s)
- Ruie Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Liewei Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology), Beijing Institute of Technology, Zhuhai 519088, China.
| | - Feng Guo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Jiawen He
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology), Beijing Institute of Technology, Zhuhai 519088, China
| | - Ka Hong Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China.
| |
Collapse
|
37
|
Liang L, Lv W, Cheng G, Gao M, Sun J, Liu N, Zhang H, Guo B, Liu J, Li Y, Xie S, Wang J, Hei J, Zhang J. Impact of celastrol on mitochondrial dynamics and proliferation in glioblastoma. BMC Cancer 2025; 25:412. [PMID: 40050778 PMCID: PMC11887396 DOI: 10.1186/s12885-025-13733-9] [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: 11/17/2024] [Accepted: 02/13/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Targeting mitochondrial dynamics offers promising strategies for treating glioblastoma multiforme. Celastrol has demonstrated therapeutic effects on various cancers, but its impact on mitochondrial dynamics in glioblastoma multiforme remains largely unknown. We studied the effects of Celastrol on mitochondrial dynamics, redox homeostasis, and the proliferation. METHODS Mito-Tracker Green staining was conducted on U251, LN229, and U87-MG cells to evaluate the effects of Celastrol on mitochondrial dynamics. The Western blot analysis quantified the expression levels of mitochondrial dynamin, antioxidant enzymes, and cell cycle-related proteins. JC-1 staining was performed to discern mitochondrial membrane potential. Mitochondrial reactive oxygen species were identified using MitoSOX. The proliferative capacity of cells was assessed using Cell Counting Kit-8 analysis, and colony formation assays. Survival analysis was employed to evaluate the therapeutic efficacy of Celastrol in C57BL/6J mice with glioblastoma. RESULTS Our findings suggest that Celastrol (1 and 1.5 µM) promotes mitochondrial fission by downregulating the expression of mitofusin-1. A decrease in mitochondrial membrane potential at 1 and 1.5 µM indicates that Celastrol impaired mitochondrial function. Concurrently, an increase in mitochondrial reactive oxygen species and impaired upregulation of antioxidant enzymes were noted at 1.5 µM, indicating that Celastrol led to an imbalance in mitochondrial redox homeostasis. At both 1 and 1.5 µM, cell proliferation was inhibited, which may be related to the decreased expression levels of Cyclin-dependent kinase 1 and Cyclin B1. Celastrol extended the survival of GBM-afflicted mice. CONCLUSION Celastrol promotes mitochondrial fission in glioblastoma multiforme cells by reducing mitofusin-1 expression, accompanying mitochondrial dysfunction, lower mitochondrial membrane potential, heightened oxidative stress, and decreased Cyclin-dependent kinase 1 and Cyclin B1 levels. This indicates that Celastrol possesses potential for repurposing as an agent targeting mitochondrial dynamics in glioblastoma multiforme, warranting further investigation.
Collapse
Affiliation(s)
- Lei Liang
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Wenying Lv
- Department of Neurosurgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Gang Cheng
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Mou Gao
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Junzhao Sun
- Department of Neurosurgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Ning Liu
- Department of Neurosurgery, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100010, China
| | - Hanbo Zhang
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Baorui Guo
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiayu Liu
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanteng Li
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | | | | | - Junru Hei
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianning Zhang
- Medical School of Chinese PLA, Beijing, 100853, China.
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
38
|
Chen Z, Han C, Xie H, Chen X, Zhang H, Sun Z, Liu M. 2-Undecanone induces ferroptosis via the STAT3/GPX4 pathway to enhance sensitivity of renal cell carcinoma to sunitinib. Biofactors 2025; 51:e70016. [PMID: 40200786 DOI: 10.1002/biof.70016] [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: 11/16/2024] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
The development of resistance significantly reduces the efficacy of targeted therapies, such as sunitinib, in renal cell carcinoma (RCC) patients, emphasizing the need for novel therapeutic agents. Natural products, known for their diverse chemical structures and mechanisms of action, offer promising anti-tumor potential with favorable safety profiles and lower toxicity compared to synthetic drugs. 2-Undecanone, a natural compound extracted from Houttuynia cordata Thunb., has demonstrated anti-tumor effects, but its specific role in RCC treatment remains unclear. In this study, we integrated network pharmacology with in vitro experiments to explore the mechanisms underlying 2-Undecanone's effects on RCC. Our results reveal that 2-Undecanone effectively inhibits RCC cell viability, proliferation, and migration. Mechanistically, we discovered that 2-Undecanone induces ferroptosis in RCC cells by promoting reactive oxygen species (ROS) generation, intracellular Fe2+ accumulation, glutathione (GSH) production, lipid peroxidation, and modulation of the STAT3/GPX4 signaling pathway. Furthermore, 2-Undecanone lowers the IC50 value of sunitinib in RCC cells, enhancing their sensitivity to this targeted therapy. Additionally, 2-Undecanone potentiates sunitinib-induced ferroptosis. In summary, our research reveals that 2-Undecanone enhances the sensitivity of RCC cells to sunitinib through targeting the STAT3/GPX4 pathway, providing new insights into potential therapeutic strategies for RCC.
Collapse
Affiliation(s)
- Zixuan Chen
- Department of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengtao Han
- Department of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiwen Xie
- Department of General Surgery, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingyu Chen
- School of Health Policy and Management, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Haojie Zhang
- Center of Structural Heart Disease, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zongrun Sun
- Department of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Liu
- Department of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Liang X, Shan X, Zhang Z, Zhou X, Shen L, Xu H, Wang Z, Redshaw C, Zhang Q. Spiropyran-based triphenylamine AlEgens: pH-responsive and light-activated mitochondria-targeted fluorescence imaging. Bioorg Chem 2025; 156:108214. [PMID: 39889554 DOI: 10.1016/j.bioorg.2025.108214] [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: 12/11/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Mitochondria are important sites of energy metabolism in eukaryotic cells and are critical for cellular physiological functions, which are closely related to mitochondrial pH (∼8.0). In this work, novel smart-responsive fluorescent probes TOCy3 and MTOCy3 for visualizing pH changes in the mitochondrial microenvironment were prepared from spiropyran structures. TOCy3 and MTOCy3 exhibited good photochromic and photoelectric switching properties. Based on the fact that Spiropyran can be exchanged between open-ring merocyanine (MC) and Spiropyran (SP) forms under the irradiation of a short-wavelength laser light source during pH change, the structure forms a semicarbocyanine structure when the structure is MC, and the molecule is positively charged and the fluorescence is enhanced. As a result, cells can be "light up", thus enhancing cellular mitochondria-targeted fluorescence imaging under light control with a Pearson's coefficient from 0.49 → 0.88. In addition, the probe is characterized by low cytotoxicity, excellent cellular uptake, and good cell migration. At the microenvironmental pH of (∼8.0) or so, it showed the highest fluorescence intensity in solution and the stability of mitochondrial targeting in living cells, indicating that TOCy3 and MTOCy3 can be applied to detect changes in subcellular organellar mitochondrial pH in solution and living cells. Thus, it can be used to monitor specific dynamic changes associated with transitions in pathological and physiological states.
Collapse
Affiliation(s)
- Xi Liang
- School of Basic Medical Sciences, Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou Medical University, Guiyang 561113, PR China
| | - Xiaofeng Shan
- School of Basic Medical Sciences, Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou Medical University, Guiyang 561113, PR China
| | - Ze Zhang
- School of Basic Medical Sciences, Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou Medical University, Guiyang 561113, PR China
| | - Xu Zhou
- School of Basic Medical Sciences, Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou Medical University, Guiyang 561113, PR China
| | - Lingyi Shen
- School of Basic Medical Sciences, Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou Medical University, Guiyang 561113, PR China
| | - Hong Xu
- School of Basic Medical Sciences, Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou Medical University, Guiyang 561113, PR China.
| | - Zhiyong Wang
- School of Basic Medical Sciences, Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou Medical University, Guiyang 561113, PR China.
| | - Carl Redshaw
- Chemistry, School of Natural Sciences, University of Hull, Hull, Yorkshire HU6 7RX, UK
| | - Qilong Zhang
- School of Basic Medical Sciences, Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou Medical University, Guiyang 561113, PR China.
| |
Collapse
|
40
|
Zhang Z, Yuan D, Jin X, Chang W, Zhang Y, Xie W. Asperosaponin VI suppresses ferroptosis in chondrocytes and ameliorates osteoarthritis by modulating the Nrf2/GPX4/HO-1 signaling pathway. Front Pharmacol 2025; 16:1539092. [PMID: 40093317 PMCID: PMC11906723 DOI: 10.3389/fphar.2025.1539092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Background Asperosaponin VI (AVI) is a naturally occurring monosaccharide derived from Dipsacus asperoides renowned for its anti-inflammatory and bone-protective properties. Objective To elucidate the specific mechanism through which AVI affects chondrocytes in osteoarthritis (OA). Methods For the in vitro experiments, primary chondrocytes were to elucidate the molecular mechanisms underlying the action of AVI.For the in vivo experiments, rat OA models were established using a modified Hulth method. The severity of knee osteoarthritis was evaluated 8 weeks post-surgery. Micro-CT imaging, hematoxylin-eosin staining, and Safranin O-fast green staining were used to assess degeneration in rat knee joints. Immunohistochemistry techniques were conducted to measure the levels of collagen II, MMP13, Nrf2, GPX4, ACSL4, and HO-1 within cartilage tissues. ELISA assays were performed to measure those of TNF-α, IL -6, and PGE2 in serum samples. Results AVI alleviated chondrocyte apoptosis and extracellular matrix degradation in rat OA induced by IL-1β. It attenuated the levels of TNF-α, IL-6, and PGE2 while reducing those of Fe2+ and malondialdehyde (MDA). AVI upregulated the expression of Nrf2, HO-1, and GPX4 while downregulating that of ACSL4. Mechanistic studies revealed that ML385-induced inhibition of the Nrf2 signaling pathway reversed the increase in GPX4 and ACSL4 expression and increased Fe2+ and MDA levels; treatment with erastin, a ferroptosis inducer, produced comparable results. In vivo experiments demonstrated that AVI improved the bone volume/tissue volume and trabecular separation values in OA rats; reversed the Osteoarthritis Research Society International score; upregulated Nrf2, HO-1, and GPX4 expression; downregulated ACSL4 and MMP13 expression, and decreased the serum levels of TNF-α, IL-6, and PGE2. Conclusion Our findings suggest that AVI is a promising therapeutic agent for OA. It exerted its protective effect by regulating the Nrf2/GPX4/HO-1 signaling axis to inhibit cartilage cell ferroptosis and improve osteoarthritis.
Collapse
Affiliation(s)
- Zhimeng Zhang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Daotong Yuan
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ximin Jin
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenjie Chang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongkui Zhang
- Department of Orthopedic Surgery, Affliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenpeng Xie
- Department of Orthopedic Surgery, Affliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
41
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [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: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
42
|
Lin YZ, Chen ZH, Yang JF, Han LJ, Yu YT, Zhan JN, Tan GC, Liu LY, Xie CL, Shan P, Jin C, Liu HX. Astaxanthin Prevents Glucocorticoid-Induced Femoral Head Osteonecrosis by Targeting Ferroptosis through the JAK2/STAT3 Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4270-4287. [PMID: 39903514 DOI: 10.1021/acs.jafc.4c09284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Glucocorticoid (GC) is extensively used in clinical practice, and the osteonecrosis of the femoral head caused by them is a common issue in orthopedic surgery, yet the underlying mechanisms remain unclear. Astaxanthin (AST), a potent natural antioxidant, has an unexplored impact on GC-induced osteonecrosis of the femoral head (GIONFH). This study explores the effects and mechanisms of AST in counteracting dexamethasone (Dex)-induced ferroptosis and GIONFH. We developed a rat model of GIONFH using intraperitoneal Dex injections and conducted in vitro analysis by culturing osteoblasts (OBs) with Dex treatment. We assessed the impact of AST on Dex-treated OBs using C11-BODIPY and FerroOrange staining, mitochondrial functionality tests, and protein expression analyses through Western blot and immunofluorescence. The influence of AST on bone microarchitecture of femoral head in rat was assessed using micro-CT, hematoxylin and eosin staining, immunofluorescence, and immunohistochemistry at imaging and histological levels. Our findings suggest that AST exerts an inhibitory effect on Dex-induced ferroptosis and GIONFH. In vitro, AST treatment increased glutathione and decreased malondialdehyde, lipid peroxidation, and mitochondrial-reactive oxygen species. Additionally, AST treatment also enhances the phosphorylation of STAT3, upregulates glutathione peroxidase 4 and osteogenic-related proteins, and stimulates bone formation. To delve deeper into the mechanism, the findings revealed that AST triggered activation of JAK2/STAT3 signaling. Moreover, the use of siRNA-STAT3 blocked the beneficial effect of AST in OBs cultivated with Dex. In brief, AST combats GIONFH by activating the JAK2/STAT3 pathway to inhibit ferroptosis.
Collapse
Affiliation(s)
- Yu-Zhe Lin
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Zi-Hao Chen
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Jian-Feng Yang
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Li-Jiang Han
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Yi-Tian Yu
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Juan-Nan Zhan
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Guang-Chan Tan
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Le-Yang Liu
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Cheng-Long Xie
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Ping Shan
- Department of Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chen Jin
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Hai-Xiao Liu
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| |
Collapse
|
43
|
Wang J, Xue J, Ma B, Zhu Y, Li J, Tian C. MEK5-ERK5 pathway mediates mitophagy by regulating Nur77 to promote tumorigenesis of osteosarcoma cells. Eur J Med Res 2025; 30:117. [PMID: 39972514 PMCID: PMC11837295 DOI: 10.1186/s40001-025-02312-0] [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: 12/10/2024] [Accepted: 01/19/2025] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVES To investigate the influence of MEK5/ERK5 pathway on mitophagy in osteosarcoma (OS), as well as the involved molecular mechanisms. METHODS The overlapped genes of mitophagy-related genes from MSigDB database and DEGs between metastatic and primary OS groups from GSE32981 were identified. GSVA of mitophagy-related pathways between the metastatic and primary groups were analyzed. The relationships between Nur77 and mitophagy-related pathways, prognosis, immune infiltrating cells, immune response gene sets were investigated. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blotting were utilized to assess the expression levels of MEK5, ERK5, Nur77, PINK1, and Parkin. Cellular behaviors and mitochondrial potential were evaluated via CCK-8, Transwell assay and JC-1 staining. RESULTS Total 4 overlapped genes were obtained as mitophagy-related DEGs, including GABARAPL1, HIF1A, PINK1, and RB1CC1. The activity scores of 3 mitophagy-related pathways exhibited significant differences between metastatic and primary groups. Importantly, Nur77 was significantly negatively correlated with a mitophagy-related pathway (GOBP MITOPHAGY: R = - 0.48, P = 0.02). The Nur77 expression in metastatic group was remarkedly higher than that in the primary group (P < 0.001). Patients with high Nur77 expression had poor prognosis, with AUC values all above 0.615 in predicting 1-, 3-, and 5-year survival. In addition, Nur77 was closely related to numerous immune cells, including activated dendritic cells, activated mast cells and M0 macrophages, and immune response gene sets chemokines and cytokines (all P < 0.05). In addition, MEK5/ERK5 pathway is activated in OS, and Nur77 is overexpressed in OS, and MEK5/ERK pathway promotes Nur77 expression, tumorigenesis and mitochondrial function in U2OS cells. Cytosporone B implement significantly increased the tumorigenesis of U2OS cells in sh-MEK5 group, and inhibited the weaken in mitochondrial membrane potential caused by MEK5 downregulation, and reversed the protein levels of mitophagy markers PINK1 and Parkin in sh-MEK5 group. CONCLUSIONS MEK5-ERK5 pathway mediates mitophagy by regulating Nur77 to promote tumorigenesis of OS cells. These findings offered promising therapeutic targets for OS.
Collapse
Affiliation(s)
- Jianshu Wang
- Department of Bone and Soft Tissue Oncology, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Jinxu Xue
- Department of Bone and Soft Tissue Oncology, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Baijing Ma
- Department of Medical Molecular Biology Research Center, Gansu Provincial Academic Institute for Medical Research, Xiaoxihu East Street, Lanzhou, 730050, China
| | - Yanqi Zhu
- Department of Bone and Soft Tissue Oncology, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Jing Li
- Department of Bone and Soft Tissue Oncology, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Caiping Tian
- Department of Medical Molecular Biology Research Center, Gansu Provincial Academic Institute for Medical Research, Xiaoxihu East Street, Lanzhou, 730050, China.
| |
Collapse
|
44
|
Liu C, Pan J, Bao Q. Ferroptosis in senescence and age-related diseases: pathogenic mechanisms and potential intervention targets. Mol Biol Rep 2025; 52:238. [PMID: 39960579 DOI: 10.1007/s11033-025-10338-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/04/2025] [Indexed: 05/09/2025]
Abstract
As the global population continues to age, the prevalence of age-related diseases is increasing, significantly influencing social and economic development, the stability of social security systems, and progress in medical technology. Ferroptosis, a recently discovered form of programmed cell death driven by iron-dependent lipid peroxidation, has emerged as a key area of research. Studies have revealed a strong association between ferroptosis and senescence. In this article, we systematically summarize the molecular mechanisms and associated signaling pathways underlying ferroptosis, emphasizing its pivotal role in the onset and progression of age-related diseases. By providing new perspectives, we aim to advance understanding of the pathogenesis of age-related diseases and guide the development of effective intervention strategies.
Collapse
Affiliation(s)
- Chang Liu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Pan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Qi Bao
- Zhejiang University School of Medicine, Hangzhou, China.
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
45
|
Han R, Luo Y, Gao J, Zhou H, Wang Y, Chen J, Zheng G, Ling C. HDAC3: A Multifaceted Modulator in Immunotherapy Sensitization. Vaccines (Basel) 2025; 13:182. [PMID: 40006729 PMCID: PMC11860249 DOI: 10.3390/vaccines13020182] [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: 12/18/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Histone deacetylase 3 (HDAC3) has emerged as a critical epigenetic regulator in tumor progression and immune modulation, positioning it as a promising target for enhancing cancer immunotherapy. This work comprehensively explores HDAC3's multifaceted roles, focusing on its regulation of key immune-modulatory pathways such as cGAS-STING, ferroptosis, and the Nrf2/HO-1 axis. These pathways are central to tumor immune evasion, antigen presentation, and immune cell activation. Additionally, the distinct effects of HDAC3 on various immune cell types-including its role in enhancing T cell activation, restoring NK cell cytotoxicity, promoting dendritic cell maturation, and modulating macrophage polarization-are thoroughly examined. These findings underscore HDAC3's capacity to reshape the tumor immune microenvironment, converting immunologically "cold tumors" into "hot tumors" and thereby increasing their responsiveness to immunotherapy. The therapeutic potential of HDAC3 inhibitors is highlighted, both as standalone agents and in combination with immune checkpoint inhibitors, to overcome resistance and improve treatment efficacy. Innovative strategies, such as the development of selective HDAC3 inhibitors, advanced nano-delivery systems, and integration with photodynamic or photothermal therapies, are proposed to enhance treatment precision and minimize toxicity. By addressing challenges such as toxicity, patient heterogeneity, and resistance mechanisms, this study provides a forward-looking perspective on the clinical application of HDAC3 inhibitors. It highlights its significant potential in personalized cancer immunotherapy, paving the way for more effective treatments and improved outcomes for cancer patients.
Collapse
Affiliation(s)
- Rui Han
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Yujun Luo
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Jingdong Gao
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
- Oncology Department, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine Suzhou, Suzhou 215009, China
| | - Huiling Zhou
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Yuqian Wang
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Jiaojiao Chen
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Guoyin Zheng
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Changquan Ling
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
46
|
Ming J, Cheng F, Fu Y, Zhang M, Rou Q, Liu K, Nuertai Z, Xu S, Tao L, Abudujapar A, Liu Y. Long non-coding RNA H19 promotes cervical cancer development via targeting the microRNA-140/ALDH1A1 axis. Eur J Med Res 2025; 30:95. [PMID: 39940029 PMCID: PMC11823256 DOI: 10.1186/s40001-025-02350-8] [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: 11/07/2024] [Accepted: 01/31/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Dysregulation of long non-coding RNA H19 (lncRNA H19) is involved in cervical cancer (CC) progression. This study aims to unveil the specific role and relevant mechanism of lncRNA H19 in CC. METHODS The expression of lncRNA H19 in CC cells was detected by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). CC cells were transfected with sh-H19, followed by cell proliferation, apoptosis, migration and invasion were examined. After location of H19 in cells using fluorescence in Situ Hybridization (FISH), target microRNAs (miRNAs) and genes associated with lncRNA H19 were predicted using bioinformatics analysis and validated by dual-luciferase reporter assay. Finally, the specific role of lncRNA H19 in CC was explored in vivo. RESULTS The upregulation of lncRNA H19 was observed in CC cells. LncRNA H19 knockdown inhibited the proliferation, migration, and invasion of CC cells, and remarkably promoted CC cell apoptosis. LncRNA H19 was localized in the nucleus and interacted with miR-140 that was downregulated in CC cells. MiR-140 inhibition reversed the effects of lncRNA H19 knockdown on CC cell development. MiR-140 targets ALDH1A1, and lncRNA H19 knockdown decreased the ALDH1A1 expression, which was rescued by miR-140 inhibition. In vivo experiments also shown that reduction of lncRNA H19 diminishes tumor growth via targeting the miR-140/ALDH1A1 axis. CONCLUSION LncRNA H19 promotes the malignant progression of CC through targeting miR-140/ALDH1A1 axis.
Collapse
Affiliation(s)
- Jie Ming
- Medical Imaging Center, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Fang Cheng
- Department of Special Needs Comprehensive, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Yating Fu
- Department of Radiology, Urumqi Stomatological Hospital, Urumqi, 830002, Xinjiang, China
| | - Meng Zhang
- Department of Special Needs Comprehensive, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Qian Rou
- Department of Special Needs Comprehensive, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Kaixiong Liu
- Department of Oncology, Bachu County People's Hospital, Bachu, 843800, Xinjiang, China
| | - Zinati Nuertai
- Department of Special Needs Comprehensive, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Shanshan Xu
- Department of Special Needs Comprehensive, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Ling Tao
- Department of Gynecology, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Alfira Abudujapar
- Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Ying Liu
- Department of Special Needs Comprehensive, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China.
- Department of Oncology, Bachu County People's Hospital, Bachu, 843800, Xinjiang, China.
| |
Collapse
|
47
|
Yang L, Tang H, Wang J, Xu D, Xuan R, Xie S, Xu P, Li X. O-GlcNAcylation attenuates ischemia-reperfusion-induced pulmonary epithelial cell ferroptosis via the Nrf2/G6PDH pathway. BMC Biol 2025; 23:32. [PMID: 39901237 PMCID: PMC11792224 DOI: 10.1186/s12915-025-02126-w] [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: 07/24/2024] [Accepted: 01/09/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Lung ischemia-reperfusion (I/R) injury is a common clinical pathology associated with high mortality. The pathophysiology of lung I/R injury involves ferroptosis and elevated protein O-GlcNAcylation levels, while the effect of O-GlcNAcylation on lung I/R injury remains unclear. This research aimed to explore the effect of O-GlcNAcylation on reducing ferroptosis in pulmonary epithelial cells caused by I/R. RESULTS First, we identified O-GlcNAc transferase 1 (Ogt1) as a differentially expressed gene in lung epithelial cells of acute lung injury/acute respiratory distress syndrome (ALI/ARDS) patients, using single-cell sequencing, and Gene Ontology analysis (GO analysis) revealed the enrichment of the ferroptosis process. We found a time-dependent dynamic alteration in lung O-GlcNAcylation during I/R injury. Proteomics analysis identified the differentially expressed proteins enriched in ferroptosis and multiple redox-related pathways based on KEGG annotation. Thus, we generated Ogt1-conditional knockout mice and found that Ogt1 deficiency aggravated ferroptosis, as evidenced by lipid reactive oxygen species (lipid ROS), malondialdehyde (MDA), Fe2+, as well as alterations in critical protein expression glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11). Consistently, we found that elevated O-GlcNAcylation inhibited ferroptosis sensitivity in hypoxia/reoxygenation (H/R) injury-induced TC-1 cells via O-GlcNAcylated NF-E2-related factor-2 (Nrf2). Furthermore, both the chromatin immunoprecipitation (ChIP) assay and the dual-luciferase reporter assay indicated that Nrf2 could bind with translation start site (TSS) of glucose-6-phosphate dehydrogenase (G6PDH) and promote its transcriptional activity. As an important rate-limiting enzyme in the pentose phosphate pathway (PPP), elevated G6PDH provided a mass of nicotinamide adenine dinucleotide phosphate (NADPH) to improve the redox state of glutathione (GSH) and eventually led to ferroptosis resistance. Rescue experiments proved that Nrf2 knockdown or Nrf2-T334A (O-GlcNAcylation site) mutation abolished the protective effect of ferroptosis resistance. CONCLUSIONS In summary, we revealed that O-GlcNAcylation could protect against I/R lung injury by reducing ferroptosis sensitivity via the Nrf2/G6PDH pathway. Our work will provide a new basis for clinical therapeutic strategies for pulmonary ischemia-reperfusion-induced acute lung injury.
Collapse
Affiliation(s)
- Liuqing Yang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China, 430071
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Hexiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Jin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China, 430071
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Dawei Xu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Rui Xuan
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
| | - Pengfei Xu
- Department of Hepatobiliary and Pancreatic Surgery, School of Pharmaceutical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| | - Xinyi Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China, 430071.
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China.
| |
Collapse
|
48
|
Yang R, Fu X, Wang Z, Xue P, Wu L, Tan X, Peng W, Li K, Gao W, Zeng P. Unlocking the potential of Traditional Chinese Medicine (TCM): Shipi Xiaoji formula (SPXJF) as a novel ferroptosis inducer in hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119236. [PMID: 39674355 DOI: 10.1016/j.jep.2024.119236] [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: 07/03/2024] [Revised: 11/07/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hepatocellular Carcinoma (HCC) is a major health concern with limited treatment options. Traditional Chinese Medicine (TCM) offers potential therapeutic approaches for HCC, and SPXJF, a TCM formula, has shown promise in clinical observations for prolonging the survival of liver cancer patients. AIM OF THE STUDY To investigate the anti-tumor effects of SPXJF on HCC cells and explore its potential mechanism, focusing on ferroptosis induction. MATERIALS AND METHODS LC/Q-TOF-MS was used for compound identification. Cell viability assays, EdU proliferation assay, colony formation assay, wound healing assay, Transwell assay, and Western-blotting were conducted to evaluate the effects of SPXJF on HCC cell proliferation, migration, and invasion. Bioinformatics analysis and RT-PCR were employed to identify potential ferroptosis-related genes and validate the results. Ferroptosis induction was investigated using ferroptosis inhibitors, ROS and lipid peroxidation detection, and TEM. In vivo experiments using a subcutaneous xenograft tumor model confirmed the anti-tumor effects of SPXJF and its ability to induce ferroptosis in HCC. RESULTS SPXJF effectively inhibited the proliferation, migration, and invasion of HCC cells in vitro. The mechanism of action was found to be related to the induction of ferroptosis, as evidenced by increased intracellular Fe2+ and ROS levels, decreased GSH levels, altered mitochondrial morphology, and upregulation of ferroptosis-inducing proteins ACSL4 and LPCAT3, along with downregulation of ferroptosis-inhibiting proteins xCT and GPX4. Bioinformatics analysis and RT-PCR further identified GSTZ1, CDC25A, AURKA, NOX4, and CAPG as potential ferroptosis-related genes regulated by SPXJF. In vivo experiments confirmed the anti-tumor effects of SPXJF and its ability to induce ferroptosis in HCC. CONCLUSIONS SPXJF exerts anti-tumor effects on HCC cells by inducing ferroptosis, and its mechanism of action involves the regulation of ferroptosis-related genes and proteins. This study provides a theoretical basis for the clinical treatment of HCC and the development of new anti-cancer drugs, offering a valuable contribution to the field of ethnopharmacology.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Animals
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Cell Proliferation/drug effects
- Mice
- Mice, Nude
- Cell Line, Tumor
- Cell Movement/drug effects
- Mice, Inbred BALB C
- Medicine, Chinese Traditional
- Xenograft Model Antitumor Assays
- Antineoplastic Agents, Phytogenic/pharmacology
- Male
- Reactive Oxygen Species/metabolism
- Cell Survival/drug effects
Collapse
Affiliation(s)
- Renyi Yang
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Cancer Research Institute of Hunan Academy of Traditional Chinese Medicine, Hunan Academy of Chinese Medicine, Hunan, 410006, China; School of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Xinying Fu
- School of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Zhibing Wang
- School of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Peisen Xue
- School of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Ling Wu
- School of Integrated Chinese and Western Medicine, Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan, 410208, China
| | - Xiaoning Tan
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Cancer Research Institute of Hunan Academy of Traditional Chinese Medicine, Hunan Academy of Chinese Medicine, Hunan, 410006, China
| | - Wei Peng
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Cancer Research Institute of Hunan Academy of Traditional Chinese Medicine, Hunan Academy of Chinese Medicine, Hunan, 410006, China
| | - Kexiong Li
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Cancer Research Institute of Hunan Academy of Traditional Chinese Medicine, Hunan Academy of Chinese Medicine, Hunan, 410006, China.
| | - Wenhui Gao
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Hunan, 410208, China.
| | - Puhua Zeng
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western, Cancer Research Institute of Hunan Academy of Traditional Chinese Medicine, Hunan Academy of Chinese Medicine, Hunan, 410006, China.
| |
Collapse
|
49
|
Wang C, You Z, Zhou G, Dong J, Tong S, Sun G. Amarogentin suppresses cell proliferation and EMT process through inducing ferroptosis in colorectal cancer. BMC Gastroenterol 2025; 25:46. [PMID: 39885392 PMCID: PMC11780999 DOI: 10.1186/s12876-025-03649-w] [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: 10/11/2024] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one common tumor with the high death rate, and badly affects the normal lives of CRC patients. Amarogentin (AG) has been found to exhibit regulatory roles and join into the progression of multiple diseases. However, the regulatory impacts and associated molecular mechanisms of AG in CRC progression keep unclear. METHODS AND RESULTS In this study, it was demonstrated that AG weakened CRC cell viability in a concentration- and time-dependent manner. In addition, AG accelerated cell apoptosis by triggering ferroptosis. The cell invasion and EMT process were restrained after AG treatment, but these impacts were reversed after Fer-1 addition. Moreover, it was uncovered that AG retarded Nrf2/HO-1/GPX4 activation. Additionally, AG modulated PTC cell viability and stimulated ferroptosis. At last, it was illustrated that AG suppressed tumor growth in vivo. CONCLUSION In conclusion, it was disclosed that AG suppressed cell proliferation and EMT process through inducing ferroptosis in CRC, and retarded Nrf2/HO-1/GPX4 activation. This discovery suggested that AG may be one effective drug for ameliorating CRC progression.
Collapse
Affiliation(s)
- Chao Wang
- Anhui Medical University, Hefei, 230022, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Zihao You
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Guoqing Zhou
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Juanjuan Dong
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Sihao Tong
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, China
| | - Guoping Sun
- Anhui Medical University, Hefei, 230022, China.
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
50
|
Zhou C, Zhong R, Zhang L, Yang R, Luo Y, Lei H, Li L, Cao J, Yuan Z, Tan X, Xie M, Qu H, He Z. Exploring the mechanism of rosmarinic acid in the treatment of lung adenocarcinoma based on bioinformatics methods and experimental validation. Discov Oncol 2025; 16:47. [PMID: 39812944 PMCID: PMC11735722 DOI: 10.1007/s12672-025-01784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVE Rosmarinic acid (RosA) is a natural polyphenol compound that has been shown to be effective in the treatment of inflammatory disease and a variety of malignant tumors. However, its specific mechanism for the treatment of lung adenocarcinoma (LUAD) has not been fully elucidated. Therefore, this study aims to clarify the mechanism of RosA in the treatment of LUAD by integrating bioinformatics, network pharmacology and in vivo experiments, and to explore the potential of the active ingredients of traditional Chinese medicine in treating LUAD. METHODS Firstly, the network pharmacology was used to screen the RosA targets, and LUAD-related differential expressed genes (DEGs) were acquired from the GEO database. The intersection of LUAD regulated by RosA (RDEGs) was obtained through the Venn diagram. Secondly, GO and KEGG enrichment analysis of RDEGs were performed, and protein-protein interaction networks (PPIs) were constructed to identify and visualize hub RDEGs. Then, molecular docking between hub RDEGs and RosA was performed, and further evaluation was carried out by using bioinformatics for the predictive value of the hub RDEGs. Finally, the mechanism of RosA in the treatment of LUAD was verified by establishing a xenograft model of NSCLC in nude mouse. RESULTS Bioinformatics and other analysis showed that, compared with the control group, the expressions of MMP-1, MMP-9, IGFBP3 and PLAU in LUAD tissues were significantly up-regulated, and the expressions of PPARG and FABP4 were significantly down-regulated, and these hub RDEGs had potential predictive value for LUAD. In vivo experimental results showed that RosA could inhibit the growth of transplanted tumors in nude mice bearing tumors of lung cancer cells, reduce the positive expression of Ki67 in lung tumor tissue, and hinder the proliferation of lung tumor cells. Upregulated expression of PPARG and FABP4 by activating the PPAR signaling pathway increases the level of ROS in lung tumor tissues and promotes apoptosis of lung tumor cells. In addition, RosA can also reduce the expression of MMP-9 and IGFBP3, inhibit the migration and invasion of lung tumor tissue cells. CONCLUSIONS This study demonstrated that RosA could induce apoptosis by regulating the PPAR signaling pathway and the expression of MMP-9, inhibit the proliferation, migration and invasion of lung cancer cells, thereby exerting anti-LUAD effects. This study provides new insight into the potential mechanism of RosA in treating LUAD and provides a new therapeutic avenue for treatment of LUAD.
Collapse
Affiliation(s)
- Chaowang Zhou
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Ruqian Zhong
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
| | - Lei Zhang
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Renyi Yang
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
| | - Yuxin Luo
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Huijun Lei
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Liang Li
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Jianzhong Cao
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Zhiying Yuan
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Xiaoning Tan
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Yuelu District, Changsha, 410006, Hunan, China
| | - Mengzhou Xie
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China.
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China.
| | - Haoyu Qu
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China.
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China.
| | - Zuomei He
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Yuelu District, Changsha, 410006, Hunan, China.
| |
Collapse
|