1
|
Liu Y, Feng LL, Han B, Cai LJ, Liu RY, Tang S, Yang Q. Exploring the molecular mechanisms through which overexpression of TET3 alleviates liver fibrosis in mice via ferroptosis in hepatic stellate cells. Cell Signal 2025; 131:111747. [PMID: 40096933 DOI: 10.1016/j.cellsig.2025.111747] [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: 12/27/2024] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
Hepatic stellate cell (HSC) activation is crucial in the onset and progression of liver fibrosis, and inhibiting or eliminating activated HSCs is a key therapeutic strategy. Ferroptosis may help eliminate activated HSCs; however, its role and regulatory pathways in liver fibrosis remain unclear. As a DNA demethylase, TET3 regulates gene expression via DNA demethylation. We previously demonstrated that TET3 overexpression alleviates CCL4-induced liver fibrosis in mice; however, the specific mechanisms, including whether TET3 affects ferroptosis in HSCs, remain unexplored. Thus, we aimed to explore the molecular mechanisms wherein TET3 overexpression improves liver fibrosis in mice via ferroptosis in HSCs. Our in vivo observations showed that overexpression of TET3 ameliorate liver fibrosis in mice, and is associated with increased levels of malondialdehyde (MDA) and Fe2+ in liver tissue, as well as decreased protein expression of SLC7A11, GPX4, and FTH1. Further in vitro studies on HSCs showed that TET3 overexpression inhibits the expression of SLC7A11, GPX4, and FTH1, and reduces intracellular GSH levels, leading to accumulation of MDA and iron ions. This induces ferroptosis in HSC-LX2 cells, while simultaneously decreasing ECM accumulation in HSCs. Furthermore, hMeDIP-SEQ and ChIP-qPCR analyses revealed that TET3 directly interacts with the promoter regions of GPX4 and FTH1 to regulate their transcriptional expression. We propose that overexpression of TET3 modulates the gene methylation status of ferroptosis-related proteins, thereby regulating HSC ferroptosis, reducing activated HSCs, and decreasing ECM deposition in the liver. This may represent one of the molecular mechanisms wherein TET3 overexpression ameliorates liver fibrosis in mice.
Collapse
Affiliation(s)
- Yin Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, 550025 Guiyang, Guizhou, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Lin-Lin Feng
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Bing Han
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, 550025 Guiyang, Guizhou, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Li-Jun Cai
- Department of Rehabilitation Medicine, The Affiliated Hospital of Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Ran-Yang Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Shuang Tang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, 550025 Guiyang, Guizhou, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Qin Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, 550025 Guiyang, Guizhou, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, 550025 Guiyang, Guizhou, China.
| |
Collapse
|
2
|
Yang G, Ren D, Yu T, Fang J. Biodegradable copper-doped calcium phosphate nanoplatform enables tumor microenvironment modulations for amplified ferroptosis in cervical carcinoma treatment. Int J Pharm X 2025; 9:100315. [PMID: 39811248 PMCID: PMC11731240 DOI: 10.1016/j.ijpx.2024.100315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
As a recently discovered form of regulated cell death, ferroptosis has attracted much attention in the field cancer therapy. However, achieving considerably enhanced efficacy is often restricted by the overexpression of endogenous glutathione (GSH) in tumor microenvironment (TME). In this work, we report a ferroptosis-inducing strategy of GSH depletion and reactive oxygen species (ROS) generation based on a biodegradable copper-doped calcium phosphate (CaP) with L-buthionine sulfoximine (BSO) loading (denoted as BSO@CuCaP-LOD, BCCL). BCCL was conducted by a biomineralization approach using lactate oxidases (LOD) as a bio-template to obtain Cu-doped CaP nanoparticles. Then, BSO was loaded to form BCCL nanoparticles with pH-responsive biodegradability to endow controlled release of Cu2+ and BSO in response to acidic TME. Benefiting from the catalytic performance of LOD, BCCL efficiently depletes the level of lactate in tumor, which can generate endogenous H2O2 for subsequent Fenton-like reaction. The Cu2+ and BSO intracellular GSH depletion followed by GSH-mediated Cu2+/Cu+ conversion, leading to the inhibition of glutathione peroxidase 4 (GPX4) and generation of •OH radicals via Cu+-mediated Fenton-like reaction. BCCL confers enhanced ferroptosis induction via intracellular LOD-induced H2O2 production, BSO-mediated GSH depletion, and Cu+-mediated ROS generation, leading to cause effective ferroptotic cell damage. As verified by in vitro and in vivo assays, the designed BCCL nanoplatform is highly biocompatible and exhibits superior anticancer therapy on uterine cervical carcinoma U14 tumor xenografts. This study, therefore, provides a biocompatible therapeutic platform that modulating the TME to enable intensive ROS generating efficacy and GSH depleting performance, as well as provides an innovative paradigm for achieving effective ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
| | | | - Tao Yu
- Department of Gynecology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, PR China
| | - Junfeng Fang
- Department of Gynecology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, PR China
| |
Collapse
|
3
|
Song C, Huang L, Li D, Zhao X. Artemisinin Alleviates Alcohol-Induced Cardiotoxicity by Inhibiting Ferroptosis via the Nrf2/NQO1 Pathway In Vivo and In Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12940-12948. [PMID: 40384105 DOI: 10.1021/acs.jafc.4c08388] [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: 05/20/2025]
Abstract
The present study was designed to explore the protective effects of artemisinin on alcohol-induced cardiac injuries and its mechanisms. In H9c2 cells, cell viability, reactive oxygen species (ROS), labile iron pool (LIP), and mitochondrial membrane potential (MMP) were measured. In the mouse model of alcohol-induced cardiomyopathy, body weight and electrocardiogram (ECG) were recorded every day. Heart tissue creatine kinase (CK), lactic dehydrogenase (LDH), iron, glutathione (GSH), malondialdehyde (MDA), and histological examination were measured. Western blot assay was performed to evaluate the expression of ferroptosis-related proteins in vitro and in vivo. The results in vitro showed that cell viability was increased, ROS and LIP contents were decreased, and the level of MMP was increased in artemisinin-treated H9c2 cells. Tissues CK, LDH, and GSH were improved after being treated with artemisinin. The ferroptosis biomarkers, including MDA and tissue iron, were attenuated after artemisinin treatment. Artemisinin protected the heart from alcohol damage by ECG and histological examination. Additionally, artemisinin down-regulated the expression of TfR and P53 and up-regulated Nrf2, HO-1, NQO1, and GPX4 expressions in vitro and in vivo. The results showed that both Fer-1 and artemisinin abolished ferroptosis. The data presented here showed that artemisinin had the potential to protect alcohol-induced cardiotoxicity through the inhibition of Nrf2/NQO1-dependent ferroptosis.
Collapse
Affiliation(s)
- Chunpu Song
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Ling Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Dongjie Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Xiaoyan Zhao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Engineering Research Center of Coptis Development & Utilization, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
4
|
Liu Y, Cai Y, Wei X, Gao K, Jin G, Zhou X, Zhao Z. Molecular Mechanisms of Potentilla Discolor Bunge in Regulating Ferroptosis to Alleviate DKD via the Nrf2 Signaling Pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025:120035. [PMID: 40414576 DOI: 10.1016/j.jep.2025.120035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 05/15/2025] [Accepted: 05/23/2025] [Indexed: 05/27/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Potentilla Discolor Bunge (PDB), a plant belonging to the Rosaceae family, is often used in traditional folk medicine to treat diabetes and prevent related complications. Additionally, fresh and tender PDB stems could be consumed as a vegetable or used to make tea. AIM OF THE STUDY To explore the potential targets and mechanisms of PDB in treating Diabetic Kidney Disease (DKD) through network pharmacology, and to investigate its role in modulating the Nrf2 signaling pathway to inhibit ferroptosis using in vivo and in vitro experiments, thereby presenting a potential therapeutic avenue for DKD. MATERIALS AND METHODS Targets of PDB compounds were screened using the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, while PDB action targets were screened using the DrugBank, OMIM, GeneCards, DisGeNET, and TTD databases. The intersecting targets were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Meanwhile, the binding modes of PDB core monomers to key targets were analysed using Molecular Docking (MD) and Surface Plasmon Resonance (SPR) experiments. Experimental validation was conducted using a High-Fat/Sucrose Diet (HFD) combined with Streptozotocin (STZ)-induced DKD rat model and an Advanced Glycation End Products (AGEs)-damaged human renal proximal tubular cell (HK-2) model. RESULTS As predicted through network pharmacology, PDB exerted therapeutic effects against DKD through multiple pathways, including AGE/RAGE, Nrf2 signaling, Oxidative Stress, and apoptosis. Furthermore, PDB's primary active constituents were Quercetin, Kaempferol, and β-sitosterol, with MD analyses suggesting strong binding affinities to the Nrf2 and Ho-1 proteins. In vivo experiments further revealed that PDB treatment reduced the 24 h urinary protein, Serum Creatinine (SCr), and urea levels. It also downregulated Malondialdehyde (MDA), Fe2+, and Reactive Oxygen Species (ROS) accumulation in renal tissues. Additionally, PDB alleviated the histopathological damage in DKD-afflicted rats and significantly upregulated Nrf2, Ho-1, Gpx4, and Slc7a11 in renal tissues. Moreover, Quercetin, Kaempferol, and β-sitosterol significantly upregulated Nrf2, Ho-1, and Gpx4, increased intracellular Glutathione (GSH) levels, reduced ROS and MDA content, and mitigated mitochondrial damage in the AGEs-exposed HK-2 cell injury model. CONCLUSION We predicted the mechanism of action of natural botanical PDB against DKD through network pharmacology, revealing that it significantly upregulated the Nrf2 signaling pathway and inhibited ferroptosis initiation, thus decelerating DKD progression. These findings were further validated through in vivo and in vitro experiments.
Collapse
Affiliation(s)
- Yunhua Liu
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yanmo Cai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyu Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kun Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ge Jin
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xin Zhou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zongjiang Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
5
|
Zheng J, Zhang W, Ito J, Henkelmann B, Xu C, Mishima E, Conrad M. N-acetyl-l-cysteine averts ferroptosis by fostering glutathione peroxidase 4. Cell Chem Biol 2025; 32:767-775.e5. [PMID: 40311609 DOI: 10.1016/j.chembiol.2025.04.002] [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: 10/02/2024] [Revised: 03/08/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
N-acetyl-l-cysteine (NAC) is a medication and a widely used antioxidant in cell death research. Despite its somewhat obscure mechanism of action, its role in inhibiting ferroptosis is gaining increasing recognition. In this study, we demonstrate that NAC treatment rapidly replenishes the intracellular cysteine pool, reinforcing its function as a prodrug for cysteine. Interestingly, its enantiomer, N-acetyl-d-cysteine (d-NAC), which cannot be converted into cysteine, also exhibits a strong anti-ferroptotic effect. We further clarify that NAC, d-NAC, and cysteine all act as direct reducing substrates for GPX4, counteracting lipid peroxidation. Consequently, only GPX4-rather than system xc-, glutathione biosynthesis, or ferroptosis suppressor protein 1-is necessary for NAC and d-NAC to prevent ferroptosis. Additionally, we identify a broad range of reducing substrates for GPX4 in vitro, including β-mercaptoethanol. These findings provide new insights into the mechanisms underlying the protective effects of NAC and other potential GPX4-reducing substrates against ferroptosis.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Weijia Zhang
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Junya Ito
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Laboratory of Food Function Analysis, Tohoku University Graduate School of Agricultural Science, Sendai, Miyagi 980-8572, Japan
| | - Bernhard Henkelmann
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Chenxi Xu
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Eikan Mishima
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8574, Japan
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, 85748 Garching, Germany.
| |
Collapse
|
6
|
Cheng W, You Y, Chen H, Zhou Y, Feng Y, Wang Y. Integrated transcriptomics and metabolomics to explore the varied hepatic toxicity induced by aged- and pristine-microplastics: in vivo and human-originated liver organoids-based in vitro study. ENVIRONMENTAL RESEARCH 2025; 280:121820. [PMID: 40378997 DOI: 10.1016/j.envres.2025.121820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/18/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025]
Abstract
Microplastics (MP) have distributed ubiquitously and emerged as a significant health risk to human beings. The adverse effect induced by aged MP at concentrations being equivalent to human internal exposure level, has raised special concern, however, is still unclear. In this study, human embryonic stem cells-derived liver organoids (LOs), a novel three-dimensional in vitro model, were exposed to 75 ng/mL self-made polypropylene (PP) and aged PP (aPP), following UV-photoaging for 0- and 500-h respectively, were subject to transcriptomic and metabolomic analysis individually and jointly, to explore the potential adverse effect of PP and aPP on human liver. The mean size of PP and aPP were 7.60 and 6.91 μm, with rough and irregular surface, and varied carbonyl index (CI) (0.08 and 0.25 respectively), indicating there were distinguished physicochemical properties. Transcriptomic analysis suggested the NADH dehydrogenase at mitochondrial complex and ATP synthesis maybe more sensitive to aPP, rather than PP. Metabolomic analysis enriched KEGG pathways including cysteine (Cys) and methionine metabolism significantly. Collectively, the homocysteine (Hcy) metabolism, were anchored upon integrated analysis. To validate, the changes in NADH dehydrogenase-encoding genes, activities of complexs, mitochondrial membrane potential, Hcy and Cys contents, as well, the cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE), were detected both in vitro and in vivo. Finally, increased serum Cys and decreased hepatic Cys were confirmed, without inflammation in the liver. The peripheral Hcy may serve as a potential biomarker for indicating the MP-induced systematic adverse health outcomes, due to the disturbance in the Hcy metabolism in the liver.
Collapse
Affiliation(s)
- Wei Cheng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yifei You
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hange Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yue Zhou
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Feng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Wang
- Ninth People's Hospital of Shanghai Jiao Tong University School of Medicine, School of Public Health, Shanghai Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Chandrasekaran K, Lee CE, Yun S, Jangid AK, Kim S, Kim K. CD44 Receptor-Mediated Ferroptosis Induction by Hyaluronic Acid Carbon Quantum Dots in Triple-Negative Breast Cancer Cells Through Downregulation of SLC7A11 Pathway. MATERIALS (BASEL, SWITZERLAND) 2025; 18:2139. [PMID: 40363642 PMCID: PMC12073876 DOI: 10.3390/ma18092139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
The field of cancer therapy is actively pursuing highly effective self-targeted drug delivery materials endowed with exceptional properties. Recently, hyaluronic acid (HA), a naturally occurring polysaccharide, has been recognized as a potential target ligand for CD44 receptors, which are frequently expressed on various solid tumor cells targeted in cancer therapy. HA carbon quantum dots (CQDs) exhibit several advantageous properties, including a high surface area-to-volume ratio, small particle size, biocompatibility, and low cytotoxicity, making them ideal for biomedical applications, such as CD44-targeted drug delivery in ferroptosis-based cancer therapy. In this study, we synthesized HA-CQDs to enhance CD44-mediated ligand-receptor interactions targeting triple-negative breast cancer (TNBC). CQDs facilitate the intracellular generation of reactive oxygen species (ROS), leading to glutathione depletion. These processes result in crucial actions such as the downregulation of glutathione peroxidase 4, downregulation of solute carrier family 7 member 11, and inhibition of cystine intake. The subsequent intracellular ROS, originating from lipid peroxidation, induces ferroptosis. Our HA-CQDs engage CD44 receptors, selectively targeting TNBCs and enhancing cancer recognition. This interaction potentially enhances the nanoplatform-based CD44 targeted therapeutic effects in inducing ferroptosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea; (K.C.); (C.E.L.); (S.Y.); (A.K.J.); (S.K.)
| |
Collapse
|
8
|
Sun F, Ma Y, Li D, Yang Q, Yuan T, Liu T, Tian X, Zhu Z, Zheng W, Wang Y, Wang W. Gentiopicroside Attenuated Dopaminergic Neurodegeneration via Inhibiting Neuroinflammatory Responses and Ferroptosis in Experimental Models of Parkinson's Disease. Basic Clin Pharmacol Toxicol 2025; 136:e70036. [PMID: 40256942 DOI: 10.1111/bcpt.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/18/2025] [Accepted: 03/23/2025] [Indexed: 04/22/2025]
Abstract
Along with the hallmark of α-synuclein deposition, neuroinflammation and iron accumulation have emerged as essential pathological features for dopaminergic neuron degeneration in PD patients and animal models. Preclinical studies have highlighted gentiopicroside's anti-inflammatory activities in treating arthritis, colitis and pancreatitis, and its neuroprotective effects on neurological diseases such as AD, chronic neuropathic pain and ischemia. However, the effects and mechanisms of gentiopicroside on PD-related conditions remain uncertain. Here, we evaluated the potential benefits of gentiopicroside using a unilateral 6-OHDA rat model and a MPP+-induced cell model. Our findings indicated that gentiopicroside improved motor deficits and restored nigral TH-positive neurons in vivo. Mechanistically, gentiopicroside ameliorated inflammatory responses of 6-OHDA-induced rats, decreased NF-κB and pro-inflammatory cytokines levels and reduced Iba-1-positive microglia in the substantia nigra. Furthermore, gentiopicroside regulated the levels of DMT1 and FPN1, thereby inhibiting iron accumulation in PD rats. In vitro, gentiopicroside preserved the viability of MPP+-treated SH-SY5Y cells and suppressed NF-κB activity and its downstream factors' levels. Meanwhile, gentiopicroside inhibited lipid peroxidation and ROS production, while it upregulated the expression of GPX4 in MPP+-treated cells. And these antiferroptosis effects were also linked to iron transporters regulation. Conclusively, gentiopicroside exhibits neuroprotective effects via alleviating neuroinflammation and iron-dependent ferroptosis, offering promise for PD treatment.
Collapse
Affiliation(s)
- Fangling Sun
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Yifu Ma
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Dan Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Qianqian Yang
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Tingwei Yuan
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Tingting Liu
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Xin Tian
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Zixin Zhu
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Wenrong Zheng
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Yufeng Wang
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Wen Wang
- Department of Laboratory Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| |
Collapse
|
9
|
Yin Y, Wang B, Yang Y, Jiang Y, Fu W. Tectorigenin mitigates homocysteine-induced inflammation and ferroptosis in BV-2 microglial cells through promoting the SIRT1/SLC7A11 pathway. Brain Res Bull 2025; 224:111272. [PMID: 40058656 DOI: 10.1016/j.brainresbull.2025.111272] [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: 10/17/2024] [Revised: 02/06/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025]
Abstract
Ferroptosis and inflammation are central to the pathophysiology of hyperhomocysteinemia (HHcy)-associated neurological disorders. Tectorigenin, a natural flavonoid aglycone extracted from numerous plants, possesses antioxidant, anti-inflammatory, and neuroprotective properties. This study aimed to investigate whether tectorigenin mitigates elevated homocysteine (Hcy)-induced toxicity in BV-2 microglial cells, focusing on its effects on inflammation and ferroptosis. Cell viability, lactate dehydrogenase (LDH) release, and proliferation assays were employed to evaluate cell injury. Inflammatory cytokines levels were determined by ELISA. Ferroptosis markers, including reactive oxygen species (ROS), lipid ROS, malondialdehyde (MDA), 4-hydroxy-nonenal (4-HNE), mitochondrial membrane potential (MMP), ATP, Fe2 + content, antioxidant enzymes (superoxide dismutase [SOD] and catalase [CAT]) activities were evaluated. mRNA and protein expressions were analyzed by qRT-PCR and western blotting, respectively. Our findings revealed that tectorigenin pretreatment significantly alleviated Hcy-induced cell injury and inflammatory response in BV-2 microglial cells. Furthermore, tectorigenin pretreatment reduced lipid peroxidation, enhanced antioxidant capacity, and alleviated ferroptotic cell death in Hcy-treated cells. Importantly, ferroptosis inhibitor Fer-1 also alleviated Hcy-induced cell injury and inflammation. Mechanistically, tectorigenin pretreatment activated the SIRT1/SLC7A11 pathway, and silencing SIRT1 reversed its protective effects. Collectively, these results indicate that tectorigenin attenuates Hcy-induced microglial injury by inhibiting inflammation and ferroptosis through the activation of the SIRT1/SLC7A11 pathway.
Collapse
Affiliation(s)
- Ye Yin
- The First Affiliated Hospital, Department of General Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Bo Wang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yan Yang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yichen Jiang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Wan Fu
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
10
|
Xu T, Ma Q, Zhang C, He X, Wang Q, Wu Y, Qin K, Liao F, Zhou P, Xu P, Yang J, Yang J, Chang J, Qiao L, Hu Y. A novel nanomedicine for osteosarcoma treatment: triggering ferroptosis through GSH depletion and inhibition for enhanced synergistic PDT/PTT therapy. J Nanobiotechnology 2025; 23:323. [PMID: 40301915 PMCID: PMC12039277 DOI: 10.1186/s12951-025-03380-4] [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: 11/12/2024] [Accepted: 04/08/2025] [Indexed: 05/01/2025] Open
Abstract
Osteosarcoma treatment remains challenging due to the limitations of single-modality therapies. To address this, we designed a carrier-free nanomedicine SRF@CuSO4.5H2O@IR780 (CSIR) for synergistic ferroptosis, photodynamic therapy (PDT), and photothermal therapy (PTT) in osteosarcoma. Interestingly, CSIR could harness the enhanced permeability and retention (EPR) effect to effectively enter tumors. Copper ions (Cu2+) within CSIR could react with the reductive intracellular environment, depleting glutathione (GSH) levels. Near-infrared (NIR) irradiation of CSIR further depleted GSH through reactive oxygen species (ROS) generation. Additionally, CSIR released sorafenib (SRF), which inhibited cystine-glutamate antiporter system xCT (xCT), thereby blocking GSH biosynthesis. RNA sequencing data confirmed ferroptosis induction by CSIR. This synergistic strategy of GSH depletion-induced ferroptosis, enhanced PDT, and photothermal cascade holds promise for improved osteosarcoma treatment and future nanomedicine design.
Collapse
Affiliation(s)
- Tangbing Xu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Heifei, 230012, China
| | - Qiming Ma
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Chi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Heifei, 230012, China
| | - Xiaoyan He
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Qian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Yunfeng Wu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Heifei, 230012, China
| | - Kunpeng Qin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Heifei, 230012, China
| | - Faxue Liao
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Heifei, 230012, China
| | - Ping Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Heifei, 230012, China
| | - Pengfei Xu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Heifei, 230012, China
| | - Jialai Yang
- Anhui Public Health Clinical Center, Heifei, 230012, China
- Department of emergency, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Junjun Yang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Heifei, 230012, China
| | - Jun Chang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Anhui Public Health Clinical Center, Heifei, 230012, China.
| | - Lei Qiao
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China.
| | - Yong Hu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
11
|
Zhang Y, Zhang D, Liao X, Xu Q, Bu L, Zheng J, Shen P, Yang C. Novel insights into the role of ferroptosis in temporomandibular joint osteoarthritis and knee osteoarthritis. Int J Med Sci 2025; 22:2119-2131. [PMID: 40303500 PMCID: PMC12035841 DOI: 10.7150/ijms.107057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by pain, limited movement, and joint stiffness, significantly impacting the quality of life and imposing substantial economic burdens. This review paper delves into the novel insights of ferroptosis, an iron-dependent form of cell death associated with lipid peroxidation, in the context of temporomandibular joint osteoarthritis (TMJ OA) and knee osteoarthritis (KOA). We explore the pathogenic characteristics of OA, including synovitis, chondrocyte death, and extracellular matrix (ECM) degradation, and discuss the limitations of current therapeutic interventions. Emerging evidence suggests a significant relationship between ferroptosis and OA, with iron accumulation and lipid peroxidation observed in osteoarthritic cartilage. This review highlights the role of ferroptosis in chondrocyte malfunction and apoptosis, inflammation, and extracellular matrix breakdown, which are central to OA pathogenesis. We also discuss potential therapeutic targets, such as Transient Receptor Potential Vanilloid 1 (TRPV1), Glutathione Peroxidase 4 (GPX4), and Nuclear Factor Erythroid 2-Related Factor 2 (NRF2), which modulate ferroptosis and OA progression. The paper consolidates studies on ferroptosis in OA, offering a comprehensive understanding of its role and the development of innovative therapies targeting this cell death mechanism to improve treatment outcomes for OA patients.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
- Shanghai Key Laboratory of Orthopedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Dahe Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Xiaoyu Liao
- Department of Rehabilitation Medicine, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215008, China
| | - Qingyu Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Lingtong Bu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Jisi Zheng
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Pei Shen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Chi Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| |
Collapse
|
12
|
Mishima E, Nakamura T, Doll S, Proneth B, Fedorova M, Pratt DA, Friedmann Angeli JP, Dixon SJ, Wahida A, Conrad M. Recommendations for robust and reproducible research on ferroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00843-2. [PMID: 40204928 DOI: 10.1038/s41580-025-00843-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
Ferroptosis is a necrotic, non-apoptotic cell death modality triggered by unrestrained iron-dependent lipid peroxidation. By unveiling the regulatory mechanisms of ferroptosis and its relevance to various diseases, research over the past decade has positioned ferroptosis as a promising therapeutic target. The rapid growth of this research field presents challenges, associated with potentially inadequate experimental approaches that may lead to misinterpretations in the assessment of ferroptosis. Typical examples include assessing whether an observed phenotype is indeed linked to ferroptosis, and selecting appropriate animal models and small-molecule modulators of ferroptotic cell death. This Expert Recommendation outlines state-of-the-art methods and tools to reliably study ferroptosis and increase the reproducibility and robustness of experimental results. We present highly validated compounds and animal models, and discuss their advantages and limitations. Furthermore, we provide an overview of the regulatory mechanisms and the best-studied players in ferroptosis regulation, such as GPX4, FSP1, SLC7A11 and ACSL4, discussing frequent pitfalls in experimental design and relevant guidance. These recommendations are intended for researchers at all levels, including those entering the expanding and exciting field of ferroptosis research.
Collapse
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Sebastian Doll
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany.
- Translational Redox Biology, TUM Natural School of Sciences, Technical University of Munich, Garching, Germany.
| |
Collapse
|
13
|
Liu S, Xu Y, Yao X, Cao H, Zhou H, Luo J, Gao H, Chen B, Chen H, Xie T, Zhan X. Perillaldehyde ameliorates sepsis-associated acute kidney injury via inhibiting HSP90AA1-mediated ferroptosis and pyroptosis: Molecular structure and protein interaction of HSP90AA1. Int J Biol Macromol 2025; 304:140954. [PMID: 39947536 DOI: 10.1016/j.ijbiomac.2025.140954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/16/2025]
Abstract
Heat shock protein 90α (HSP90AA1) is a molecular chaperone involved in a variety of cellular processes. Special attention is paid to how perillaldehyde ameliorates kidney injury by inhibiting HSP90AA1-mediated iron and pyrotoxicity, and in-depth analysis of the molecular structure and protein interactions of HSP90AA-1. The interaction between perillaldehyde and HSP90AA1 and the effect of perillaldehyde on the molecular structure of HSP90AA1 were analyzed by molecular docking and surface plasmon resonance technique. Western blot and immunohistochemical results showed that perillaldehyde could decrease the expression of HSP90AA1 and change its distribution in the kidney. Molecular docking and surface plasmonic resonance experiments revealed the high affinity binding between perillaldehyde and HSP90AA1, and further analysis showed that perillaldehyde could induce the conformational change of HSP90AA1, thereby inhibiting its function.
Collapse
Affiliation(s)
- Shuai Liu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yunfei Xu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Heng Cao
- Department of Urology, The Third the People's Hospital of Bengbu, Bengbu Medical College, Bengbu 233000, China
| | - Hongmin Zhou
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Jun Luo
- Department of Urology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Hanlu Gao
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Bowen Chen
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Hao Chen
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Tiancheng Xie
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Xiangcheng Zhan
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
14
|
Zheng J, Conrad M. Ferroptosis: when metabolism meets cell death. Physiol Rev 2025; 105:651-706. [PMID: 39661331 DOI: 10.1152/physrev.00031.2024] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
We present here a comprehensive update on recent advancements in the field of ferroptosis, with a particular emphasis on its metabolic underpinnings and physiological impacts. After briefly introducing landmark studies that have helped to shape the concept of ferroptosis as a distinct form of cell death, we critically evaluate the key metabolic determinants involved in its regulation. These include the metabolism of essential trace elements such as selenium and iron; amino acids such as cyst(e)ine, methionine, glutamine/glutamate, and tryptophan; and carbohydrates, covering glycolysis, the citric acid cycle, the electron transport chain, and the pentose phosphate pathway. We also delve into the mevalonate pathway and subsequent cholesterol biosynthesis, including intermediate metabolites like dimethylallyl pyrophosphate, squalene, coenzyme Q (CoQ), vitamin K, and 7-dehydrocholesterol, as well as fatty acid and phospholipid metabolism, including the biosynthesis and remodeling of ester and ether phospholipids and lipid peroxidation. Next, we highlight major ferroptosis surveillance systems, specifically the cyst(e)ine/glutathione/glutathione peroxidase 4 axis, the NAD(P)H/ferroptosis suppressor protein 1/CoQ/vitamin K system, and the guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin/dihydrofolate reductase axis. We also discuss other potential anti- and proferroptotic systems, including glutathione S-transferase P1, peroxiredoxin 6, dihydroorotate dehydrogenase, glycerol-3-phosphate dehydrogenase 2, vitamin K epoxide reductase complex subunit 1 like 1, nitric oxide, and acyl-CoA synthetase long-chain family member 4. Finally, we explore ferroptosis's physiological roles in aging, tumor suppression, and infection control, its pathological implications in tissue ischemia-reperfusion injury and neurodegeneration, and its potential therapeutic applications in cancer treatment. Existing drugs and compounds that may regulate ferroptosis in vivo are enumerated.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, Garching, Germany
| |
Collapse
|
15
|
Li Y, Li A, Xing Y, Jiang Y, Pei H, He Z, Li J, Zhao Y, Shi K, Zong Y, Du R. Gastrodin targets the system Xc-/GPX4 axis to inhibit abnormal proliferation of fibroblast-like synoviocytes and improve rheumatoid arthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156493. [PMID: 39970857 DOI: 10.1016/j.phymed.2025.156493] [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: 10/25/2024] [Revised: 01/30/2025] [Accepted: 02/09/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Rheumatoid Arthritis (RA) is a chronic autoimmune disease characterized by the abnormal proliferation of Fibroblast-like Synoviocytes (FLS), leading to synovial hyperplasia and progressive joint destruction in an inflammatory environment. Gastrodin (GAS), the main active ingredient of Gastrodia elata, is also the basis for its anti-inflammatory, anti-tumor, and other pharmacological effects. However, GAS's effects on RA and its specific molecular mechanisms remain to be thoroughly explored. PURPOSE This study combines transcriptomics and modern pharmacological methods to explore the molecular mechanism of GAS to ameliorate RA and provide a theoretical basis for the development of new therapeutic strategies for rheumatoid arthritis. METHODS A rat adjuvant arthritis (AA) model was established to further determine the molecular mechanism by which GAS affects RA based on transcriptomics and molecular docking. The pharmacological activity of GAS against RA was evaluated by in vivo experiments such as micro-CT, histopathological examination, transmission electron microscopy and Elisa. Primary FLS cells were also extracted from AA rats for in vitro experiments to reveal the potential of GAS in treating RA. RESULTS Transcriptomic and molecular docking analyses suggested ferroptosis as a potential mechanism for GAS to ameliorate RA. In vitro and in vivo studies demonstrated that GAS downregulates SLC7A11, impacts the antioxidant system, and inhibits GPX4. It facilitates lipopolysaccharide-induced ferroptosis in RA-FLS (rheumatoid arthritis fibroblast-like synoviocytes) cells, suppresses synovial proliferation, and ameliorates rheumatoid arthritis. CONCLUSION This study reveals the molecular mechanism of the anti-RA action of GAS from a novel perspective, suggesting that GAS is a promising drug candidate for the treatment of RA.
Collapse
Affiliation(s)
- Yanlu Li
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China
| | - Aoyun Li
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China
| | - Yanchao Xing
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China
| | - Yingshan Jiang
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China
| | - Hongyan Pei
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China
| | - Zhongmei He
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun Jilin Province, 130118, PR China
| | - Jianming Li
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun Jilin Province, 130118, PR China
| | - Yan Zhao
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun Jilin Province, 130118, PR China
| | - Kun Shi
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun Jilin Province, 130118, PR China
| | - Ying Zong
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun Jilin Province, 130118, PR China.
| | - Rui Du
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, Jilin Province, 130118, China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun Jilin Province, 130118, PR China; Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, Jilin Province, 130118, China.
| |
Collapse
|
16
|
Fu J, Liu H, Liang Y, Shi Y, Gao X, Chen P, Yu D, Wang Y, Lu F, Liu S. Study on the mechanism of Huangqi Chifeng decoction regulating ferroptosis inhibiting smooth muscle cells derived foam cell formation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119507. [PMID: 39978445 DOI: 10.1016/j.jep.2025.119507] [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: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese medicine, specifically Huangqi Chifeng Decoction (HQCF), is recognized for its efficacy in treating atherosclerosis (AS), a common cardiovascular disease. Despite its established benefits in addressing Qi deficiency, blood stasis, and collateral obstruction, the precise mechanism through which HQCF affects AS remains unclear. AIM OF THE STUDY This study investigated the potential of HQCF to mitigate AS by suppressing smooth muscle cell (SMC) foam formation through the ferroptosis pathway. MATERIALS AND METHODS An AS model was established using ApoE-/- mice fed a high-fat diet (HFD), and the role of HQCF in regulating ferroptosis in AS was examined. Using a single-cell proteomics analysis strategy, we identified the primary targets of HQCF in SMCs. Additionally, an oxidized low-density lipoprotein (ox-LDL)-treated SMC-derived foam cell model was established. The effects HQCF on SMC ferroptosis were analyzed, and ox-LDL-induced SMCs were pretreated with small interfering RNA (siRNA) and overexpressing carrier plasmids (pcDNA) to identify potential therapeutic targets, for specifically thioredoxin (TXN). RESULTS HQCF the pathological state of the aorta in ApoE-/- mice, regulated lipid levels, improved antioxidant capacity, modulated the phenotypic transformation of SMCs, and maintained the dynamic balance of extracellular matrix degradation and remodeling. Additionally, HQCF may inhibit ferroptosis via positive regulation of the GPX4/xCT signaling pathway. Single-cell proteomics revealed 36 common differentially expressed proteins (DEPs), suggesting that HQCF's treatment of AS may be associated with the regulation of cellular function and redox homeostasis. The abnormal expression of TXN in SMCs may be related to the phenotypic transition induced by AS. HQCF was also found to ameliorate oxidative stress and mitochondrial dysfunction during SMC foaming. Moreover, ferroptosis was involved in ox-LDL-induced foam cell formation, and HQCF alleviated these pathologies by inhibiting ferroptosis. The protective effect of HQCF on SMCs was enhanced by TXN overexpression but partially reversed by TXN knockdown, further indicating that HQCF's regulation of SMC function and inhibition of ferroptosis is, at least in part, mediated by TXN. CONCLUSION These findings suggest that HQCF protects SMCs from ferroptosis by regulating the TXN/xCT/GPX4 pathway, ameliorating the aortic pathological state, alleviating oxidative stress, and maintaining mitochondrial homeostasis in mice.
Collapse
MESH Headings
- Animals
- Ferroptosis/drug effects
- Drugs, Chinese Herbal/pharmacology
- Foam Cells/drug effects
- Foam Cells/metabolism
- Foam Cells/pathology
- Atherosclerosis/drug therapy
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Male
- Mice
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Mice, Inbred C57BL
- Diet, High-Fat
- Lipoproteins, LDL
- Thioredoxins/metabolism
- Thioredoxins/genetics
- Disease Models, Animal
- Mice, Knockout, ApoE
- Mice, Knockout
Collapse
Affiliation(s)
- Jiaqi Fu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongyu Liu
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yuqin Liang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yunhe Shi
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xin Gao
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Pingping Chen
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Donghua Yu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yu Wang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Shumin Liu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
17
|
Lin J, Li B, Guo X, Li G, Zhang Q, Wang W. Key Mechanisms of Oxidative Stress-Induced Ferroptosis in Heart Failure with Preserved Ejection Fraction and Potential Therapeutic Approaches. Rev Cardiovasc Med 2025; 26:26613. [PMID: 40160560 PMCID: PMC11951494 DOI: 10.31083/rcm26613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025] Open
Abstract
The prevalence of heart failure with preserved ejection fraction (HFpEF) is increasing annually, particularly among patients with metabolic disorders such as hypertension and diabetes. However, there is currently no treatment capable of altering the natural course of HFpEF. Recently, the interplay between oxidative stress and ferroptosis in cardiovascular diseases has drawn extensive attention; however, minimal research has been published on the mechanisms of oxidative stress and ferroptosis in HFpEF. This paper reviews the relevant mechanisms through which oxidative stress is induced and promotes ferroptosis during the development of HFpEF. The review also explores more efficacious treatment approaches for HFpEF by inhibiting oxidative stress and ferroptosis, thereby offering a theoretical foundation for verifying the feasibility of these methods for further research. As tumor-targeted therapy progresses, the survival period of tumor patients is prolonged, and cardiovascular events have gradually emerged as one of the most crucial causes of death among tumor patients. Hence, inhibiting the vascular endothelial growth factor (VEGF) pathway has become a major target in tumor treatment, significantly enhancing patient survival. Nevertheless, secondary cardiovascular complications and events, such as myocardial injury and subsequent heart failure, have severely impacted patient survival and quality of life. Therefore we have also explored the potential mechanism through which novel targeted anti-cancer drugs induce HFpEF via ferroptosis. Additionally, we reviewed the specific modes of action for preventing and treating HFpEF without influencing their anti-cancer therapeutic effect.
Collapse
Affiliation(s)
- Junling Lin
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Bingtao Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Xueqi Guo
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Guodong Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Qi Zhang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Wenjuan Wang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| |
Collapse
|
18
|
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: 1] [Impact Index Per Article: 1.0] [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
|
19
|
Solak K, Yildiz Arslan S, Acar M, Turhan F, Unver Y, Mavi A. Combination of magnetic hyperthermia and gene therapy for breast cancer. Apoptosis 2025; 30:99-116. [PMID: 39427089 DOI: 10.1007/s10495-024-02026-4] [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] [Accepted: 10/01/2024] [Indexed: 10/21/2024]
Abstract
This study presented a novel breast cancer therapy model that uses magnetic field-controlled heating to trigger gene expression in cancer cells. We created silica- and amine-modified superparamagnetic nanoparticles (MSNP-NH2) to carry genes and release heat under an alternating current (AC) magnetic field. The heat-inducible expression plasmid (pHSP-Azu) was designed to encode anti-cancer azurin and was delivered by magnetofection. MCF-7 cells demonstrated over 93% cell viability and 12% transfection efficiency when exposed to 75 µg/ml of MSNP-NH2, 3 µg of DNA, and PEI at a 0.75 PEI/DNA ratio (w: w), unlike non-tumorigenic cells (MCF-10 A). Magnetic hyperthermia (MHT) increased azurin expression by heat induction, leading to cell death in dual ways. The combination of MHT and heat-regulated azurin expression induced cell death, specifically in cancer cells, while having negligible effects on MCF-10 A cells. The proposed strategy clearly shows that simultaneous use of MHT and MHT-induced azurin gene expression may selectively target and kill cancer cells, offering a promising direction for cancer therapy.
Collapse
Affiliation(s)
- Kubra Solak
- Department of Nanoscience and Nanoengineering, Institute of Science and Technology, Atatürk University, Erzurum, Türkiye
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Türkiye
| | - Seyda Yildiz Arslan
- Department of Molecular Biology and Genetics, Institute of Science and Technology, Atatürk University, Erzurum, Türkiye
| | - Melek Acar
- Department of Molecular Biology and Genetics, Institute of Science and Technology, Atatürk University, Erzurum, Türkiye
| | - Fatma Turhan
- Department of Molecular Biology and Genetics, Institute of Science and Technology, Atatürk University, Erzurum, Türkiye
| | - Yagmur Unver
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Türkiye.
| | - Ahmet Mavi
- Department of Nanoscience and Nanoengineering, Institute of Science and Technology, Atatürk University, Erzurum, Türkiye.
- Department of Mathematics and Science Education, Education Faculty of Kazim Karabekir, Atatürk University, Erzurum, Türkiye.
| |
Collapse
|
20
|
Li X, Zhou Z, Tao Y, He L, Zhan F, Li J. Linking homocysteine and ferroptosis in cardiovascular disease: insights and implications. Apoptosis 2024; 29:1944-1958. [PMID: 39044092 DOI: 10.1007/s10495-024-01999-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 07/25/2024]
Abstract
Homocysteine (Hcy) is a metabolic intermediate product derived from methionine. Hyperhomocysteinemia is a condition associated with various diseases. Hcy is recognized as a risk factor for cardiovascular disease (CVD). Ferroptosis, a novel form of cell death, is primarily characterized by substantial iron accumulation and lipid peroxidation. Recent research indicates a close association between ferroptosis and the pathophysiological processes of tumors, neurological diseases, CVD, and other ailments. However, limited research has been conducted on the impact of Hcy on ferroptosis. Therefore, this paper aimed to investigate the potential roles and mechanisms of homocysteine and ferroptosis in the context of cardiovascular disease. By conducting comprehensive literature research and analysis, we aimed to summarize recent advancements in understanding the effects of homocysteine on ferroptosis in cardiovascular diseases. This research contributes to a profound understanding of this critical domain.
Collapse
Affiliation(s)
- Xiaozhong Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China
| | - Zheng Zhou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China
| | - Yu Tao
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lei He
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Fenfang Zhan
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
21
|
Wu Y, Miao Y, Cao Y, Gong Z. Gastrodin prevents myocardial injury in sleep-deprived mice by suppressing ferroptosis through SIRT6. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9111-9121. [PMID: 38896272 DOI: 10.1007/s00210-024-03230-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Gastrodin (GAS), a bioactive compound derived from the orchid plant Gastrodia elata, exhibits numerous pharmacological effects. However, its effect on sleep deprivation (SD)-induced cardiac injury and the mechanisms are unknown. This study established SD mice model using a modified multiple platform water method and induced ferroptosis model in H9c2 cells using Erastin. The heart rate of mice was measured, and myocardial and mitochondrial structures were visualized using hematoxylin and eosin (H&E) staining and transmission electron microscopy (TEM). Myocardial injury, oxidative stress indicators, and Fe2+ levels were detected by the kit method. The reactive oxygen species (ROS) levels were detected by immunofluorescence, and SIRT6 and ferroptosis-associated protein expression levels were detected by Western blot. Reduced heart rate and abnormalities in myocardial tissue and mitochondrial structure were ameliorated in the SD group of mice after GAS treatment. GAS treatment reduced ROS levels in Erastin-induced H9c2 cells. GAS treatment reduced atrial natriuretic peptide (ANP), creatine kinase (CK), lactate dehydrogenase (LDH), malondialdehyde (MAD), and Fe2+ levels, and increased superoxide dismutase (SOD) and glutathione (GSH) levels in the SD and Erastin groups. Western blot showed that GAS treatment increased the expression of sirtuin 6 (SIRT6), solute carrier family 7 member 11 (SLC7A11), and glutathione peroxidase 4 (GPX4) and decreased the expression of P53 in SD and Erastin groups. The SIRT6 inhibitor OSS_128167 (OSS) reversed GAS treatment of Erastin-induced ferroptosis in H9c2 cells. These observations propose that GAS prevents myocardial injury in sleep-deprived mice by suppressing ferroptosis through SIRT6.
Collapse
Affiliation(s)
- Yi Wu
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China
| | - Yuping Miao
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China
| | - Yin Cao
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China.
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang, 550014, Guizhou Province, China.
| | - Zipeng Gong
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang, 550014, Guizhou Province, China.
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
22
|
Wang LY, Zhang L, Bai XY, Qiang RR, Zhang N, Hu QQ, Cheng JZ, Yang YL, Xiang Y. The Role of Ferroptosis in Amyotrophic Lateral Sclerosis Treatment. Neurochem Res 2024; 49:2653-2667. [PMID: 38864944 DOI: 10.1007/s11064-024-04194-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: 03/25/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rare neurodegenerative disease with a challenging treatment landscape, due to its complex pathogenesis and limited availability of clinical drugs. Ferroptosis, an iron-dependent form of programmed cell death (PCD), stands distinct from apoptosis, necrosis, autophagy, and other cell death mechanisms. Recent studies have increasingly highlighted the role of iron deposition, reactive oxygen species (ROS) accumulation, oxidative stress, as well as systemic Xc- and glutamate accumulation in the antioxidant system in the pathogenesis of amyotrophic lateral sclerosis. Therefore, targeting ferroptosis emerges as a promising strategy for amyotrophic lateral sclerosis treatment. This review introduces the regulatory mechanism of ferroptosis, the relationship between amyotrophic lateral sclerosis and ferroptosis, and the drugs used in the clinic, then discusses the current status of amyotrophic lateral sclerosis treatment, hoping to provide new directions and targets for its treatment.
Collapse
Affiliation(s)
- Le Yi Wang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xin Yue Bai
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Rong Rong Qiang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Ning Zhang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qian Qian Hu
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Jun Zhi Cheng
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Yan Ling Yang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Yang Xiang
- College of Physical Education, Yan'an University, Shaanxi, 716000, China.
| |
Collapse
|
23
|
Shi JX, Zhang ZC, Yin HZ, Piao XJ, Liu CH, Liu QJ, Zhang JC, Zhou WX, Liu FC, Yang F, Wang YF, Liu H. RNA m6A modification in ferroptosis: implications for advancing tumor immunotherapy. Mol Cancer 2024; 23:213. [PMID: 39342168 PMCID: PMC11437708 DOI: 10.1186/s12943-024-02132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
The pursuit of innovative therapeutic strategies in oncology remains imperative, given the persistent global impact of cancer as a leading cause of mortality. Immunotherapy is regarded as one of the most promising techniques for systemic cancer therapies among the several therapeutic options available. Nevertheless, limited immune response rates and immune resistance urge us on an augmentation for therapeutic efficacy rather than sticking to conventional approaches. Ferroptosis, a novel reprogrammed cell death, is tightly correlated with the tumor immune environment and interferes with cancer progression. Highly mutant or metastasis-prone tumor cells are more susceptible to iron-dependent nonapoptotic cell death. Consequently, ferroptosis-induction therapies hold the promise of overcoming resistance to conventional treatments. The most prevalent post-transcriptional modification, RNA m6A modification, regulates the metabolic processes of targeted RNAs and is involved in numerous physiological and pathological processes. Aberrant m6A modification influences cell susceptibility to ferroptosis, as well as the expression of immune checkpoints. Clarifying the regulation of m6A modification on ferroptosis and its significance in tumor cell response will provide a distinct method for finding potential targets to enhance the effectiveness of immunotherapy. In this review, we comprehensively summarized regulatory characteristics of RNA m6A modification on ferroptosis and discussed the role of RNA m6A-mediated ferroptosis on immunotherapy, aiming to enhance the effectiveness of ferroptosis-sensitive immunotherapy as a treatment for immune-resistant malignancies.
Collapse
Affiliation(s)
- Jun-Xiao Shi
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Zhi-Chao Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Hao-Zan Yin
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Xian-Jie Piao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Cheng-Hu Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Qian-Jia Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Jia-Cheng Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Wen-Xuan Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu-Chen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu Yang
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China.
- Key Laboratory of Biosafety Defense, Ministry of Education, Shanghai, 200433, China.
- Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China.
| | - Yue-Fan Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| |
Collapse
|
24
|
Guan M, Yu Q, Zhou G, Wang Y, Yu J, Yang W, Li Z. Mechanisms of chondrocyte cell death in osteoarthritis: implications for disease progression and treatment. J Orthop Surg Res 2024; 19:550. [PMID: 39252111 PMCID: PMC11382417 DOI: 10.1186/s13018-024-05055-6] [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: 08/18/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by the degeneration, destruction, and excessive ossification of articular cartilage. The prevalence of OA is rising annually, concomitant with the aging global population and increasing rates of obesity. This condition imposes a substantial and escalating burden on individual health, healthcare systems, and broader social and economic frameworks. The etiology of OA is multifaceted and not fully understood. Current research suggests that the death of chondrocytes, encompassing mechanisms such as cellular apoptosis, pyroptosis, autophagy, ferroptosis and cuproptosis, contributes to both the initiation and progression of the disease. These cell death pathways not only diminish the population of chondrocytes but also exacerbate joint damage through the induction of inflammation and other deleterious processes. This paper delineates the morphological characteristics associated with various modes of cell death and summarizes current research results on the molecular mechanisms of different cell death patterns in OA. The objective is to review the advancements in understanding chondrocyte cell death in OA, thereby offering novel insights for potential clinical interventions.
Collapse
Affiliation(s)
- Mengqi Guan
- Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Qingyuan Yu
- Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Guohui Zhou
- Orthopedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Yan Wang
- Sino-Japanese Friendship Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Jianan Yu
- Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Wei Yang
- Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Zhenhua Li
- Orthopedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China.
| |
Collapse
|
25
|
Li S, Zhang G, Hu J, Tian Y, Fu X. Ferroptosis at the nexus of metabolism and metabolic diseases. Theranostics 2024; 14:5826-5852. [PMID: 39346540 PMCID: PMC11426249 DOI: 10.7150/thno.100080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Ferroptosis, an iron-dependent form of regulated cell death, is emerging as a crucial regulator of human physiology and pathology. Increasing evidence showcases a reciprocal relationship between ferroptosis and dysregulated metabolism, propagating a pathogenic vicious cycle that exacerbates pathology and human diseases, particularly metabolic disorders. Consequently, there is a rapidly growing interest in developing ferroptosis-based therapeutics. Therefore, a comprehensive understanding of the intricate interplay between ferroptosis and metabolism could provide an invaluable resource for mechanistic insight and therapeutic development. In this review, we summarize the important metabolic substances and associated pathways in ferroptosis initiation and progression, outline the cascade responses of ferroptosis in disease development, overview the roles and mechanisms of ferroptosis in metabolic diseases, introduce the methods for ferroptosis detection, and discuss the therapeutic perspectives of ferroptosis, which collectively aim to illustrate a comprehensive view of ferroptosis in basic, translational, and clinical science.
Collapse
Affiliation(s)
- Shuangwen Li
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Guixiang Zhang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiankun Hu
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Tian
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xianghui Fu
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
26
|
Nakamura T, Conrad M. Exploiting ferroptosis vulnerabilities in cancer. Nat Cell Biol 2024; 26:1407-1419. [PMID: 38858502 DOI: 10.1038/s41556-024-01425-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/17/2024] [Indexed: 06/12/2024]
Abstract
Ferroptosis is a distinct lipid peroxidation-dependent form of necrotic cell death. This process has been increasingly contemplated as a new target for cancer therapy because of an intrinsic or acquired ferroptosis vulnerability in difficult-to-treat cancers and tumour microenvironments. Here we review recent advances in our understanding of the molecular mechanisms that underlie ferroptosis and highlight available tools for the modulation of ferroptosis sensitivity in cancer cells and communication with immune cells within the tumour microenvironment. We further discuss how these new insights into ferroptosis-activating pathways can become new armouries in the fight against cancer.
Collapse
Affiliation(s)
- Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets & Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets & Therapeutics Center, Helmholtz Munich, Neuherberg, Germany.
| |
Collapse
|
27
|
Zhang M, Zhong J, Song Z, Xu Q, Chen Y, Zhang Z. Regulatory mechanisms and potential therapeutic targets in precancerous lesions of gastric cancer: A comprehensive review. Biomed Pharmacother 2024; 177:117068. [PMID: 39018877 DOI: 10.1016/j.biopha.2024.117068] [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: 05/10/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024] Open
Abstract
Precancerous lesions of gastric cancer (PLGC) represent a critical pathological stage in the transformation from normal gastric mucosa to gastric cancer (GC). The global incidence of PLGC has been rising over the past few decades, with a trend towards younger onset ages. Increasing evidence suggests that early prevention and treatment of PLGC can effectively reverse the malignant development of gastric mucosal epithelial cells. However, there is currently a lack of effective therapeutic drugs and methods. Recent years have witnessed substantial advancements in PLGC research, with the elucidation of novel regulatory mechanisms offering promising avenues for clinical intervention and drug development. This review aims to delineate potential targets for early prevention and diagnosis of GC while exploring innovative approaches to PLGC management. This article focuses on elucidating the regulatory mechanisms of the inflammatory microenvironment, bile acids (BA), glycolysis, autophagy, apoptosis, ferroptosis, and cellular senescence. We pay particular attention to potential therapeutic targets for PLGC, with the goal of providing insights and theoretical basis for clinical research on PLGC.
Collapse
Affiliation(s)
- Maofu Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Jialin Zhong
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhongyang Song
- Department of Oncology, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730020, China
| | - Qian Xu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yuchan Chen
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhiming Zhang
- Department of Oncology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu 730050, China.
| |
Collapse
|
28
|
Zeng H, Zhang Y, Liu N, Wei Q, Yang F, Li J. Stimulus-Responsive Nanodelivery and Release Systems for Cancer Gene Therapy: Efficacy Improvement Strategies. Int J Nanomedicine 2024; 19:7099-7121. [PMID: 39045344 PMCID: PMC11265383 DOI: 10.2147/ijn.s470637] [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: 03/25/2024] [Accepted: 06/18/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction of exogenous genes into target cells to overcome various tumor diseases caused by genetic defects or abnormalities and gene therapy, a new treatment method, provides a promising strategy for tumor treatment. Over the past decade, gene therapy has made exciting progress; however, it still faces the challenge of low nucleic acid delivery and release efficiencies. The emergence of nonviral vectors, primarily nanodelivery and release systems (NDRS), has resulted in a historic breakthrough in the application of gene therapy. NDRS, especially stimulus-responsive NDRS that can respond in a timely manner to changes in the internal and external microenvironment (eg, low pH, high concentration of glutathione/reactive oxygen species, overexpressed enzymes, temperature, light, ultrasound, and magnetic field), has shown excellent loading and release advantages in the precision and efficiency of tumor gene therapy and has been widely applied. The only disadvantage is that poor transfection efficiency limits the in-depth application of gene therapy in clinical practice, owing to the presence of biological barriers in the body. Therefore, this review first introduces the development history of gene therapy, the current obstacles faced by gene delivery, strategies to overcome these obstacles, and conventional vectors, and then focuses on the latest research progress in various stimulus-responsive NDRS for improving gene delivery efficiency. Finally, the future challenges and prospects that stimulus-responsive NDRS may face in clinical application and transformation are discussed to provide references for enhancing in-depth research on tumor gene therapy.
Collapse
Affiliation(s)
- Huamin Zeng
- Cancer Prevention and Treatment Institute of Chengdu, Department of Pathology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan, 611130, People’s Republic of China
| | - Yiran Zhang
- Cancer Prevention and Treatment Institute of Chengdu, Department of Pathology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan, 611130, People’s Republic of China
- School of Medical and Life Sciences, Chengdu University of traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Ningyi Liu
- Cancer Prevention and Treatment Institute of Chengdu, Department of Pathology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan, 611130, People’s Republic of China
- School of Basic Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637007, People’s Republic of China
| | - Qingqing Wei
- Cancer Prevention and Treatment Institute of Chengdu, Department of Pathology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan, 611130, People’s Republic of China
- School of Basic Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637007, People’s Republic of China
| | - Fan Yang
- Cancer Prevention and Treatment Institute of Chengdu, Department of Pathology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan, 611130, People’s Republic of China
| | - Jie Li
- Institute of Herbgenomics, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| |
Collapse
|
29
|
Yu Y, Liang J, Yuan Z, Wang A, Liu X, Chen Y, Zhang M, Gao Y, Zhang H, Liu Y. Bioactive compound schaftoside from Clinacanthus nutans attenuates acute liver injury by inhibiting ferroptosis through activation the Nrf2/GPX4 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118135. [PMID: 38556139 DOI: 10.1016/j.jep.2024.118135] [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: 01/22/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Clinacanthus nutans (Burm. f.) Lindau, a traditional herb renowned for its anti-tumor, antioxidant, and anti-inflammatory properties, has garnered considerable attention. Although its hepatoprotective effects have been described, there is still limited knowledge of its treatment of acute liver injury (ALI), and its mechanisms remain unclear. AIM OF THE STUDY To assess the efficacy of Clinacanthus nutans in ALI and to identify the most effective fractions and their underlying mechanism of action. METHODS Bioinformatics was employed to explore the underlying anti-hepatic injury mechanisms and active compounds of Clinacanthus nutans. The binding ability of schaftoside, a potential active ingredient in Clinacanthus nutans, to the core target nuclear factor E2-related factor 2 (Nrf2) was further determined by molecular docking. The role of schaftoside in improving histological abnormalities in the liver was observed by H&E and Masson's staining in an ALI model induced by CCl4. Serum and liver biochemical parameters were measured using AST, ALT and hydroxyproline kits. An Fe2+ kit, transmission electron microscopy, western blotting, RT-qPCR, and DCFH-DA were used to measure whether schaftoside reduces ferroptosis-induced ALI. Subsequently, specific siRNA knockdown of Nrf2 in AML12 cells was performed to further elucidate the mechanism by which schaftoside attenuates ferroptosis-induced ALI. RESULTS Bioinformatics analysis and molecular docking showed that schaftoside is the principal compound from Clinacanthus nutans. Schaftoside was shown to diminish oxidative stress levels, attenuate liver fibrosis, and forestall ferroptosis. Deeper investigations revealed that schaftoside amplified Nrf2 expression and triggered the Nrf2/GPX4 pathway, thereby reversing mitochondrial aberrations triggered by lipid peroxidation, GPX4 depletion, and ferroptosis. CONCLUSION The lead compound schaftoside counters ferroptosis through the Nrf2/GPX4 axis, providing insights into a novel molecular mechanism for treating ALI, thereby presenting an innovative therapeutic strategy for ferroptosis-induced ALI.
Collapse
Affiliation(s)
- Yi Yu
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Jingwei Liang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Haikou, 571199, China
| | - Zhexin Yuan
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Aiping Wang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Xinxing Liu
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Yu Chen
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Min Zhang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Yanan Gao
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Haikou, 571199, China
| | - Haiying Zhang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Haikou, 571199, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, Haikou, 571199, China.
| | - Yan Liu
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Haikou, 571199, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, Haikou, 571199, China.
| |
Collapse
|
30
|
Song C, Li D, Huang L, Zhang J, Zhao X. Role of Ferroptosis Regulation by Nrf2/NQO1 Pathway in Alcohol-Induced Cardiotoxicity In Vitro and In Vivo. Chem Res Toxicol 2024; 37:1044-1052. [PMID: 38833663 DOI: 10.1021/acs.chemrestox.4c00140] [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: 06/06/2024]
Abstract
The aim of the present study was to evaluate the cardiotoxic effects of alcohol and its potential toxic mechanism on ferroptosis in mice and H9c2 cells. Mice were intragastrically treated with three different concentrations of alcohol, 7, 14, and 28%, each day for 14 days. Body weight and electrocardiography (ECG) were recorded over the 14 day period. Serum creatine kinase (CK), lactic dehydrogenase (LDH), MDA, tissue iron, and GSH levels were measured. Cardiac tissues were examined histologically, and ferroptosis was assessed. In H9c2 cardiomyocytes, cell viability, reactive oxygen species (ROS), labile iron pool (LIP), and mitochondrial membrane potential (MMP) were measured. The proteins of ferroptosis were evaluated by the western blot technique in vivo and in vitro. The results showed that serum CK, LDH, MDA, and tissue iron levels significantly increased in the alcohol treatment group in a dose-dependent manner. The content of GSH decreased after alcohol treatment. ECG and histological examinations showed that alcohol impaired cardiac function and structure. In addition, the levels of ROS and LIP increased, and MMP levels decreased after alcohol treatment. Ferrostatin-1 (Fer-1) protected cells from lipid peroxidation. Western blotting analysis showed that alcohol downregulated the expression of Nrf2, NQO1, HO-1, and GPX4. The expressions of P53 and TfR were upregulated in vivo and in vitro. Fer-1 significantly alleviated alcohol-induced ferroptosis. In conclusion, the study showed that Nrf2/NQO1-dependent ferroptosis played a vital role in the cardiotoxicity induced by alcohol.
Collapse
Affiliation(s)
- Chunpu Song
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Dongjie Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Ling Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Jie Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Xiaoyan Zhao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
31
|
Mehmood AH, Chang J, Wang Y, Li S, Ma J, Dong B, Liu H. A D-π-A-type ratiometric fluorescent probe to detect polarity changes and inhibition effect during ferroptosis. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:3486-3491. [PMID: 38804096 DOI: 10.1039/d4ay00519h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
To thoroughly understand ferroptosis's biological functions in living cells, it is crucial to investigate the polarity variations that occur during this unique Fe(II)-facilitated oxidative type of cell death. In this work, we report the development of a ratiometric probe (Po-P) to visualize the polarity changes in living cells and the inhibition effect during ferroptosis. The polarity-responsive fluorophore utilized by Po-P has a D-π-A-type structure. Based on theoretical calculations, ICT was proposed as the basis for Po-P's polarity-responsive mechanism. According to cell imaging results, Po-P had a desirable capacity for monitoring polarity fluctuations and erastin-induced ferroptosis. Furthermore, inhibition imaging revealed that dihydrolipoic acid (DHLA) could potentially prevent polarity changes that occur during erastin-induced ferroptosis, just as vitamin E (VE). We anticipate that the probe Po-P could be a valuable tool to quickly monitor polarity fluctuations and inhibition effects during ferroptosis and create new medications for treating disorders related to ferroptosis.
Collapse
Affiliation(s)
- Abdul Hadi Mehmood
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Jia Chang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Yan Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Shijing Li
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Jiale Ma
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Baoli Dong
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Hong Liu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| |
Collapse
|
32
|
Wu L, Xue X, He C, Lai Y, Tong L. Cell death‑related molecules and targets in the progression of urolithiasis (Review). Int J Mol Med 2024; 53:52. [PMID: 38666544 PMCID: PMC11090264 DOI: 10.3892/ijmm.2024.5376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
Urolithiasis is a high‑incidence disease caused by calcium oxalate (mainly), uric acid, calcium phosphate, struvite, apatite, cystine and other stones. The development of kidney stones is closely related to renal tubule cell damage and crystal adhesion and aggregation. Cell death, comprising the core steps of cell damage, can be classified into various types (i.e., apoptosis, ferroptosis, necroptosis and pyroptosis). Different crystal types, concentrations, morphologies and sizes cause tubular cell damage via the regulation of different forms of cell death. Oxidative stress caused by high oxalate or crystal concentrations is considered to be a precursor to a variety of types of cell death. In addition, complex crosstalk exists among numerous signaling pathways and their key molecules in various types of cell death. Urolithiasis is considered a metabolic disorder, and tricarboxylic acid cycle‑related molecules, such as citrate and succinate, are closely related to cell death and the inhibition of stone development. However, a literature review of the associations between kidney stone development, metabolism and various types of cell death is currently lacking, at least to the best of our knowledge. Thus, the present review summarizes the major advances in the understanding of regulated cell death and urolithiasis progression.
Collapse
Affiliation(s)
- Liping Wu
- Department of Pharmacy, Ganzhou People's Hospital, Ganzhou, Jiangxi 341099, P.R. China
| | - Xiaoyan Xue
- Department of Pharmacy, Ganzhou People's Hospital, Ganzhou, Jiangxi 341099, P.R. China
| | - Chengwu He
- Department of Urology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, P.R. China
| | - Yongchang Lai
- Department of Urology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, P.R. China
- Department of Pharmaceutical Management, School of Medical Business, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Lingfei Tong
- Department of Pharmacy, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
33
|
Jiang YP, Liu BG, Dang Y, Liu LJ, Pang Y, Bai XD, Sun F, Kang TH, Zhao ZH. Integrative analysis of transcriptomics and metabolomics reveals the protective effect and mechanism of salidroside on testicular ischemia-reperfusion injury. Front Pharmacol 2024; 15:1377836. [PMID: 38818379 PMCID: PMC11137215 DOI: 10.3389/fphar.2024.1377836] [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: 02/21/2024] [Accepted: 03/26/2024] [Indexed: 06/01/2024] Open
Abstract
Testicular torsion is a critical urologic condition for which testicular detorsion surgery is considered irreplaceable as well as the golden method of reversal. However, the surgical treatment is equivalent to a blood reperfusion process, and no specific drugs are available to treat blood reperfusion injuries. Salidroside (SAL) is one of the main effective substances in rhodiola, which has been shown to have antioxidant and antiapoptosis activities. This study was designed to determine whether SAL exerted a protective effect on testicular ischemia-reperfusion (I/R) injury. In this study, the I/R injury model of the testes and reoxygenation (OGD/R) model were used for verification, and SAL was administered at doses of 100 mg/kg and 0.05 mmol/L, respectively. After the experiments, the testicular tissue and TM4 Sertoli cells were collected for histopathologic and biochemical analyses. The results revealed that SAL improves the structure of testicular tissue and regulates the oxidation-antioxidation system. To further understand the molecular mechanisms of SAL in treating testicular I/R injuries, transcriptomics and metabonomics analyses were integrated. The results show that the Nfr2/HO-1/GPX4/ferroptosis signaling pathway is enriched significantly, indicating that it may be the main regulatory pathway for SAL in the treatment of testicular I/R injuries. Thereafter, transfection with Nrf2 plasmid-liposome was used to reverse verify that the Nfr2/HO-1/GPX4/ferroptosis signaling pathway was the main pathway for SAL anti-testicular I/R injury treatment. Thus, it is suggested that SAL can protect against testicular I/R injuries by regulating the Nfr2/HO-1/GPX4 signaling pathway to inhibit ferroptosis and that SAL may be a potential drug for the treatment of testicular I/R injuries.
Collapse
Affiliation(s)
- Ya Ping Jiang
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Shaanxi, China
- Department of Pharmacy, Xianyang Hospital of Yan’an University, Shaanxi, China
| | - Bao Gui Liu
- Department of Pharmacy, Xianyang Hospital of Yan’an University, Shaanxi, China
| | - Yi Dang
- Department of Clinical Research, Xianyang Hospital of Yan’an University, Shaanxi, China
| | - Lin Jie Liu
- Department of Pharmacy, Xianyang Hospital of Yan’an University, Shaanxi, China
| | - Yang Pang
- Department of Pharmacy, Xianyang Hospital of Yan’an University, Shaanxi, China
| | - Xiao Dong Bai
- Department of Clinical Research, Xianyang Hospital of Yan’an University, Shaanxi, China
| | - Feng Sun
- Department of Science and Education, Xianyang Hospital of Yan’an University, Shaanxi, China
| | - Tian Hong Kang
- Department of Pharmacy, Xianyang Hospital of Yan’an University, Shaanxi, China
| | - Zheng Hang Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Shaanxi, China
| |
Collapse
|