1
|
Zheng Q, Wang D, Lin R, Xu W. Pyroptosis, ferroptosis, and autophagy in spinal cord injury: regulatory mechanisms and therapeutic targets. Neural Regen Res 2025; 20:2787-2806. [PMID: 39101602 PMCID: PMC11826477 DOI: 10.4103/nrr.nrr-d-24-00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/24/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Regulated cell death is a form of cell death that is actively controlled by biomolecules. Several studies have shown that regulated cell death plays a key role after spinal cord injury. Pyroptosis and ferroptosis are newly discovered types of regulated cell deaths that have been shown to exacerbate inflammation and lead to cell death in damaged spinal cords. Autophagy, a complex form of cell death that is interconnected with various regulated cell death mechanisms, has garnered significant attention in the study of spinal cord injury. This injury triggers not only cell death but also cellular survival responses. Multiple signaling pathways play pivotal roles in influencing the processes of both deterioration and repair in spinal cord injury by regulating pyroptosis, ferroptosis, and autophagy. Therefore, this review aims to comprehensively examine the mechanisms underlying regulated cell deaths, the signaling pathways that modulate these mechanisms, and the potential therapeutic targets for spinal cord injury. Our analysis suggests that targeting the common regulatory signaling pathways of different regulated cell deaths could be a promising strategy to promote cell survival and enhance the repair of spinal cord injury. Moreover, a holistic approach that incorporates multiple regulated cell deaths and their regulatory pathways presents a promising multi-target therapeutic strategy for the management of spinal cord injury.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Weihong Xu
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
2
|
Mavaddatiyan L, Naeini S, Khodabandeh S, Hosseini F, Skelton RP, Azizi V, Talkhabi M. Exploring the association between aging, ferroptosis, and common age-related diseases. Arch Gerontol Geriatr 2025; 135:105877. [PMID: 40339241 DOI: 10.1016/j.archger.2025.105877] [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: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
Aging is a natural biological process that is characterized by the progressive decline in physiological functions and an increased vulnerability to age-related diseases. The aging process is driven by different cell and molecular mechanisms. It has recently been shown that aging is associated with heightened vulnerability to ferroptosis (an intracellular iron-dependent form of programmed cell death). This susceptibility arises from various factors including oxidative stress, impaired antioxidant defences, and dysregulated iron homeostasis. The progressive decline in cellular antioxidant capacity and the accumulation of damaged components contribute to the increased susceptibility of aging cells to ferroptosis. Dysregulation of key regulators involved in ferroptosis, such as glutathione peroxidase 4 (GPX4), iron regulatory proteins, and lipid metabolism enzymes, further exacerbates this vulnerability. The decline in cellular defence mechanisms against ferroptosis during aging contributes to the accumulation of damaged cells and tissues, ultimately resulting in the manifestation of age-related diseases. Understanding the intricate relevance between aging and ferroptosis holds significant potential for developing strategies to counteract the detrimental effects of aging and age-related diseases. This will subsequently act to mitigate the negative consequences of aging and improving overall health in the elderly population. This review aims to clarify the relationship between aging and ferroptosis, and explores the underlying mechanisms and implications for age-related disorders, including neurodegenerative, cardiovascular, and neoplastic diseases. We also discuss the accumulating evidence suggesting that the imbalance of redox homeostasis and perturbations in iron metabolism contribute to the age-associated vulnerability to ferroptosis.
Collapse
Affiliation(s)
- Laleh Mavaddatiyan
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - SaghiHakimi Naeini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Sara Khodabandeh
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Hosseini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - RhysJ P Skelton
- Flinders Medical Centre, Department of Ophthalmology, Bedford Park, Australia
| | - Vahid Azizi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mahmood Talkhabi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
3
|
Zhang L, Wang H, Liang B, Qin L, Zhang M, Lv X, Hu S, Fan X, Xie W, Yang H, Huang G, Jing W, Zhao J. Ponicidin promotes ferroptosis to enhance treatment sensitivity in Lenvatinib-resistant hepatocellular carcinoma cells through regulation of KEAP1/NRF2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156824. [PMID: 40382941 DOI: 10.1016/j.phymed.2025.156824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/17/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE This study explores the therapeutic potential of Ponicidin on Lenvatinib-resistant hepatocellular carcinoma (HCC), elucidates its mechanism in reversing Lenvatinib resistance, and provides experimental evidence for its clinical application in overcoming this resistance. METHODS Huh7 and HCC-LM3 cells were used to construct Lenvatinib-resistant cell lines, Huh7-LR and HCC-LM3-LR. Changes in the ferroptosis pathway post-drug resistance were observed by measuring ferroptosis-related markers. The proliferation assay were assessed by CCK-8, while the migration and invasion were measured by scratch and Transwell invasion assays. In mechanistic study, chip analysis and immunoprecipitation with biotin-labeled Ponicidin, were conducted to explore how Ponicidin overcame drug resistance. Xenograft model in nude mice was established to examine Ponicidin's anti-HCC effects In vivo. Clinical specimens were used to assess the true status of patients in Lenvatinib-resistant HCC patients. RESULTS Our study reveals for the first time that ferroptosis inhibition drives Lenvatinib resistance in HCC and identifies Ponicidin as a novel KEAP1-targeting agent to reverse this process. In vitro, ferroptosis pathway was suppressed in Lenvatinib-resistant cells. Ponicidin suppressed proliferation, clonogenicity, migration, and invasion in these cells. The combination of Ponicidin and Lenvatinib significantly inhibited proliferation and reversed drug resistance by activating the ferroptosis pathway. Preliminary mechanistic studies showed that Ponicidin binds to KEAP1, stabilizing the KEAP1/NRF2 interaction, inhibiting the nuclear translocation and activation of NRF2, and thereby inducing ferroptosis to overcome Lenvatinib resistance. In vivo, the combination of Ponicidin and Lenvatinib exhibited a synergistic effect, significantly delaying tumor growth. Clinically, p-NRF2 and GPX4 expression was higher in the Lenvatinib-insensitive group, suggesting that the ferroptosis pathway was inhibited in these patients. Thus, this study demonstrated that Ponicidin promotes ferroptosis to enhances treatment sensitivity in Lenvatinib-resistant HCC cells through KEAP1/NRF2.
Collapse
Affiliation(s)
- Lisha Zhang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China; Nanchang Hongdu Hospital of TCM, Nanchang, PR China
| | - Hao Wang
- Department of Oncology, The Air Force Hosptital of Northern Theater PLA, Shenyang, China
| | - Beibei Liang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Lijuan Qin
- Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Mingzhu Zhang
- University of Shanghai for Science and Technology, Shanghai, PR China
| | - Xingxian Lv
- Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Shi Hu
- Department of Biomedical Engineering, College of Basic Medical Sciences, Second Military Medical University, Shanghai, PR China
| | - Xiaoyu Fan
- Department of Molecular Biology, Shanghai Center for Clinical Laboratory, Shanghai, PR China
| | - Wei Xie
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Wei Jing
- Department of Surgery, Changhai Hospital, Second Military Medical University, Shanghai, PR China.
| | - Jian Zhao
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China.
| |
Collapse
|
4
|
Zhang X, Liu J, Bai C, Fan Y, Song H, Huang Z, Li Y, Luo T. Palmitic acid enhances the sensitivity of ferroptosis via endoplasmic reticulum stress mediated the ATF4/TXNIP Axis in polycystic ovary syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156777. [PMID: 40393214 DOI: 10.1016/j.phymed.2025.156777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/27/2025] [Accepted: 04/10/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Palmitic acid (PA), the most prevalent saturated fatty acid in humans, is closely associated with ovarian dysfunction. Elevated PA levels in the follicular fluid of patients with polycystic ovary syndrome (PCOS) are correlated with the outcomes of assisted reproductive technology (ART), though the underlying mechanism remains unclear. METHODS Multi-omics analysis identified PA and TXNIP as potential pathogenic factors in PCOS. CCK-8 and apoptosis assay were conducted to detect the cytotoxicity of PA. To further investigate the molecular mechanisms underlying PA-induced ferroptosis, we established COV434 cell models with both TXNIP overexpression and knockdown. Transmission electron microscopy (TEM), western blot (WB), ELISA assays, and flow cytometry were employed to assess ferroptosis-related markers. A PCOS mouse model was also developed, and histopathological staining, TEM, ELISA, and WB were performed to evaluate clinical parameters related to PCOS ovarian ferroptosis levels. To clarify the targeting relationship between ATF4 and TXNIP, we utilized luciferase reporter gene assays, chromatin immunoprecipitation (ChIP), and RT-qPCR for a comprehensive analysis. RESULTS In vivo and in vitro, PA enhanced the sensitivity of PCOS ovarian ferroptosis. The protein levels of TXNIP and ACSL4 were upregulated in both PCOS patients and mouse models after PA treatment. PA also induces the expression of the ferroptosis inhibitor SLC7A11 as part of an adaptive response. Elevated intracellular ROS levels, increased MDA content, decreased GSH/GSSG ratios, elevated ferrous iron levels, and TEM findings collectively indicated that PA induces ferroptosis in KGN/COV434 cells. The ER stress inhibitor 4-PBA reduces PA-induced ferroptosis in PCOS ovaries by suppressing ER stress, thereby improving PA-induced PCOS-like traits. Moreover, the UPR gene ATF4 regulates cellular ferroptosis by activating the transcriptional expression of TXNIP. CONCLUSION PA stimulated ovarian ferroptosis by activating ER stress, a process mediated by the ATF4/TXNIP axis, which might represent a potential mechanism underlying the progression of PCOS. The application of ER stress inhibitors improved PCOS traits by reducing the sensitivity of ovarian ferroptosis.
Collapse
Affiliation(s)
- Xumin Zhang
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Jianrong Liu
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China; Department of reproductive Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China.
| | - Chunmei Bai
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Yanxin Fan
- Department of Orthopedics, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, 29 Shuangtasi Street, Taiyuan 030012, Shanxi, PR China
| | - Haixia Song
- Department of reproductive Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Ziwei Huang
- Department of reproductive Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Yang Li
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| | - Ting Luo
- The Fifth Clinical Medical College of Shanxi Medical University, 29 Shuangtaisi street, Taiyuan 030012, Shanxi, PR China
| |
Collapse
|
5
|
Lin L, Huang D, Huang H, Xie L, Huang Y, Ye C, Chu L, Qiao Y, Meng X, Cai S, Dong H. Lung microbiota metabolite L-malic acid attenuates the airway inflammation in asthma by inhibiting ferroptosis. Toxicol Appl Pharmacol 2025; 500:117396. [PMID: 40381742 DOI: 10.1016/j.taap.2025.117396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Inhaled environmental allergens, such as house dust mites (HDM), have been shown to induce an inflammatory reaction, tissue injury, and increased airway sensitivity in the lungs, ultimately leading to the development of allergic asthma. The imbalance of respiratory microbiota and metabolites plays a crucial role in the progression of allergic asthma. However, there is limited knowledge available regarding the alterations in respiratory microbiota and metabolites and their impact on the host in the context of asthma. The aim of this study was to investigate the potential pathways involved in the development of asthma through the analysis of lung flora and metabolites. A mouse model of house dust mite (HDM)-induced asthma was established, and alveolar lavage samples were collected for microbiome 16S rRNA sequencing and untargeted metabolic analysis. Microbiological analyses indicated a significant alteration in the microbiota after 4 and 6 weeks of HDM nebulisation stimulation. This was characterized by a decrease in microbial diversity, as well as reductions in the relative proportion of Gallionella and Lactobacillus. Conversely, the abundance of Flavobacterium and Ralstonia increased in the HDM4W and HDM6W groups, respectively. Metabolomic analyses revealed seven distinct metabolites, among them L-malic acid, which were linked to signaling pathways in a mouse model of HDM-induced asthma. The correlation analysis demonstrated a positive association between L-malic acid and Rhodanobacter and Nocardioides. L-malic acid was discovered to be efficacious in reducing airway inflammation in mice with house dust mite-induced asthma. Further analysis revealed that this change was linked to lipid peroxidation and changes in ferroptosis markers, namely GPX4 and FTH. These findings suggest that L-malate inhibits ferroptosis. However, the introduction of ferroptosis inducers, such as Erastin, was observed to negate the beneficial effect of butyrate. In summary, this research implies that the respiratory microbiota metabolite L-malic acid lessens airway inflammation in asthma by inhibiting ferroptosis, offering a potential approach for managing asthma.
Collapse
Affiliation(s)
- Lishan Lin
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Respiratory and Critical Care Medicine of Puning People's Hospital, Puning, China
| | - Danhui Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haohua Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lingyan Xie
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cuiping Ye
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lanhe Chu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yujie Qiao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaojing Meng
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Yao R, Zeng Y, Zhang Y, Cao X, Mao J, Li W, Xu K, Liu L. Identification of a new micropeptide altKLF4 derived from KLF4 that influences myeloma chemotherapeutic sensitivity. Cell Signal 2025; 131:111767. [PMID: 40147548 DOI: 10.1016/j.cellsig.2025.111767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Multiple myeloma (MM) is a common yet incurable hematological malignancy characterized by bone marrow infiltration. A major clinical challenge is the resistance to chemotherapy, highlighting the urgent need to better understand the molecular mechanisms underlying chemotherapeutic resistance to available drugs. Recent studies have emphasized the role of micropeptides in solid tumors and leukemia, but their functions in MM remain unclear. In this study, we identified a novel micropeptide, altKLF4, derived from the transcription factor KLF4, which is highly expressed in newly diagnosed myeloma patient samples. We found that ectopic expression of altKLF4 interfered with chemotherapy sensitivity induced by proteasome inhibitors in myeloma cells. Additionally, confocal microscopy and transcriptome sequencing revealed that altKLF4 co-localizes with the mitochondrial inner marker TOMM20 and participates in mitochondria-related biological processes, suggesting that altKLF4 partially localizes to the mitochondria. Mitochondria may also play a role in regulating ferroptosis. Our results further demonstrated that altKLF4 inhibited drug sensitivity and ferroptosis induced by the GPX4 inhibitor RSL3 in multiple myeloma cells through a direct interaction with GPX4. In vivo experiments showed that RSL3 significantly suppressed primary myeloma growth, which could be rescued by the micropeptide altKLF4. Taken together, our study identifies altKLF4 as a novel micropeptide that serves as a potential biomarker for chemotherapeutic resistance in multiple myeloma, offering insights for diagnosis and management of drug-resistant MM.
Collapse
Affiliation(s)
- Ruosi Yao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yindi Zeng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yaxin Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Cao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiwei Mao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenjing Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Linlin Liu
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
7
|
Guo W, Duan Z, Wu J, Zhou BP. Epithelial-mesenchymal transition promotes metabolic reprogramming to suppress ferroptosis. Semin Cancer Biol 2025; 112:20-35. [PMID: 40058616 DOI: 10.1016/j.semcancer.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular de-differentiation process that provides cells with the increased plasticity and stem cell-like traits required during embryonic development, tissue remodeling, wound healing and metastasis. Morphologically, EMT confers tumor cells with fibroblast-like properties that lead to the rearrangement of cytoskeleton (loss of stiffness) and decrease of membrane rigidity by incorporating high level of poly-unsaturated fatty acids (PUFA) in their phospholipid membrane. Although large amounts of PUFA in membrane reduces rigidity and offers capabilities for tumor cells with the unbridled ability to stretch, bend and twist in metastasis, these PUFA are highly susceptible to lipid peroxidation, which leads to the breakdown of membrane integrity and, ultimately results in ferroptosis. To escape the ferroptotic risk, EMT also triggers the rewiring of metabolic program, particularly in lipid metabolism, to enforce the epigenetic regulation of EMT and mitigate the potential damages from ferroptosis. Thus, the interplay among EMT, lipid metabolism, and ferroptosis highlights a new layer of intricated regulation in cancer biology and metastasis. Here we summarize the latest findings and discuss these mutual interactions. Finally, we provide perspectives of how these interplays contribute to cellular plasticity and ferroptosis resistance in metastatic tumor cells that can be explored for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Wenzheng Guo
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Zhibing Duan
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Jingjing Wu
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Binhua P Zhou
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
8
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
9
|
Gu W, Pan T, Wang X, Kang L, Liu N, Piao M, Feng C. Sevoflurane exposure triggers ferroptosis of neuronal cells initiated by the activation of ATM/p53 in the neonatal mouse brain via JNK/p38 MAPK-mediated oxidative DNA damage. Int Immunopharmacol 2025; 158:114866. [PMID: 40378436 DOI: 10.1016/j.intimp.2025.114866] [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: 03/06/2025] [Revised: 04/27/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025]
Abstract
Neuronal death has long been regarded as a pivotal pathological factor in the developmental neurotoxicity caused by the volatile anesthetic sevoflurane in the neonatal brain, but the detailed mechanism remains controversial. Ferroptosis is a novel type of regulated cell death driven by excess lipid peroxidation secondary to intracellular iron overload, and it is implicated in the pathogenesis of various neurological disorders. Acting as a death messenger, p53 is primarily activated by ATM during DNA damage and mediates various forms of cell death, including apoptosis, autophagy, and ferroptosis. JNK/p38 MAPK are important stress-responsive pathways that can exacerbate intracellular ROS production, thereby linking DNA damage to many pathological conditions such as neurodegeneration and ischemic injury. In our present study, we demonstrated that sevoflurane exposure-induced neuronal death was correlated with intracellular iron overload and lipid peroxidation in HT22 cells, primary hippocampal neurons, and the hippocampi of neonatal mice, consistent with the hallmarks of ferroptosis. Furthermore, we found that sevoflurane-induced neuronal ferroptosis was associated with ATM/p53 activation in response to DNA damage. Additionally, sevoflurane exposure caused JNK/p38 MAPK activation followed by intracellular ROS accumulation, ultimately leading to DNA damage. Mechanistically, ATM/p53 contributed to ferroptosis caused by sevoflurane via two pathways: (1) enhancing iron uptake (upregulating TFR and downregulating FPN) and (2) promoting lipid peroxidation through NOX4, ALOX12, ALOX15 activation and SLC7A11 suppression. Collectively, these findings demonstrated that sevoflurane exposure induced ferroptosis of neuronal cells in the neonatal brain, triggered by ATM/p53 activation via JNK/p38 MAPK-mediated ROS accumulation and subsequent DNA damage.
Collapse
Affiliation(s)
- Wanping Gu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Tingting Pan
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Xuedong Wang
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Liheng Kang
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Nan Liu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Meihua Piao
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China.
| | - Chunsheng Feng
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China.
| |
Collapse
|
10
|
Jing H, Wang GH, Yang K, Zhu ZS, Wang Y, Chen ZY, Sun N, Zhang M. Immunometabolic interplay and molecular characterization of hepcidins reveal ferroptosis mechanisms to Edwardsiella piscicida infection in black rockfish Sebastes schlegeli. Int J Biol Macromol 2025; 318:145037. [PMID: 40484093 DOI: 10.1016/j.ijbiomac.2025.145037] [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: 03/19/2025] [Revised: 05/15/2025] [Accepted: 06/05/2025] [Indexed: 06/16/2025]
Abstract
Edwardsiella piscicida infection represents a major pathogenic threat in aquaculture, yet the molecular mechanisms underlying host-pathogen interactions is still not fully understood. Here, we investigated the pathophysiological response of black rockfish Sebastes schlegeli to E. piscicida infection through an integrated approach combining immune-metabolism analysis and molecular characterization. Infected fish displayed severe pathological manifestations, including melanin deposition, visceral swelling, and extensive hepatic damage, with mortality reaching 80 % by day 15 post-infection. Transcriptomic analysis identified 5363 differentially expressed genes, while metabolomic profiling revealed 194 altered metabolites. Integration of these datasets demonstrated significant perturbations in key metabolic pathways, including glutathione metabolism, nucleotide metabolism, and energy metabolism, highlighting the host's metabolic reprogramming in response to infection. Notably, we identified ferroptosis as a key mechanism of E. piscicida-induced tissue damage, characterized by glutathione depletion and dysregulation of iron homeostasis genes. Furthermore, we characterized two hepcidin genes (Sshepcidin 1 and Sshepcidin 2) that exhibited tissue-specific expression patterns and were significantly upregulated following bacterial challenge. Synthetic mature peptides demonstrated broad-spectrum antibacterial activity against various bacterial pathogens, with mSshep 1 exhibiting greater overall potency than mSshep 2. These findings provide novel insights into the molecular underpinnings of black rockfish response to E. piscicida infection and highlight potential targets for therapeutic intervention in aquaculture systems.
Collapse
Affiliation(s)
- Hao Jing
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266237, China
| | - Guang-Hua Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266237, China
| | - Kai Yang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266237, China
| | - Zhi-Shu Zhu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266237, China
| | - Yue Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266237, China
| | - Zi-Yue Chen
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266237, China
| | - Nuo Sun
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266237, China
| | - Min Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266237, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, Shandong Province, 266237, China.
| |
Collapse
|
11
|
Tang K, Ye T, He Y, Ba X, Xia D, Peng E, Chen Z, Ye Z, Yang X. Ferroptosis, necroptosis, and pyroptosis in calcium oxalate crystal-induced kidney injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167791. [PMID: 40086520 DOI: 10.1016/j.bbadis.2025.167791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Kidney stones represent a highly prevalent urological disorder worldwide, with high incidence and recurrence rates. Calcium oxalate (CaOx) crystal-induced kidney injury serves as the foundational mechanism for the formation and progression of CaOx stones. Regulated cell death (RCD) such as ferroptosis, necroptosis, and pyroptosis are essential in the pathophysiological process of kidney injury. Ferroptosis, a newly discovered RCD, is characterized by its reliance on iron-mediated lipid peroxidation. Necroptosis, a widely studied programmed necrosis, initiates with a necrotic phenotype that resembles apoptosis in appearance. Pyroptosis, a type of RCD that involves the gasdermin protein, is accompanied by inflammation and immune response. In recent years, increasing amounts of evidence has demonstrated that ferroptosis, necroptosis, and pyroptosis are significant pathophysiological processes involved in CaOx crystal-induced kidney injury. Herein, we summed up the roles of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury. Furthermore, we delved into the curative potential of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury.
Collapse
Affiliation(s)
- Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Ye
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Yen CC, Chen PCH, Chen SC, Wu WC, Yen CH, Lin YC, Wu PK, Chen CM, Wang JY, Chao TC, Yang MH, Fletcher JA. Ferroptosis as a therapeutic vulnerability in MDM2 inhibition in dedifferentiated liposarcoma. Oncol Lett 2025; 29:269. [PMID: 40247991 PMCID: PMC12005077 DOI: 10.3892/ol.2025.15015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Ferroptosis is a form of necrotic cell death characterized by phospholipid oxidation. The cystine-glutamate antiporter (xCT), composed of solute carrier family 7 member 11 (SLC7A11) and SLC3A2, imports cystine for glutathione synthesis. Glutathione peroxidase 4 (GPX4) requires glutathione to counteract lipid peroxidation and prevent ferroptosis. Erastin, an xCT inhibitor, and Ras-selective lethal small molecule 3 (RSL3), a GPX4 inhibitor, suppress GPX4 function and induce ferroptosis. Tumor protein p53 (TP53) has a paradoxical role in ferroptosis regulation. Mouse double minute 2 homolog (MDM2), a negative regulator of TP53, is a key oncogene in well-differentiated liposarcoma (WDLPS) and dedifferentiated liposarcoma (DDLPS). Therefore, the present study explored the role of ferroptosis in DDLPS treatment response and resistance. Publicly available expression profiles of WDLPS, DDLPS and adipose tissue were analyzed, and the differential expression of ferroptosis-related genes regulated by the MDM2-TP53 pathway was identified in WDLPS and DDLPS. In vitro experiments were performed to assess the effects of erastin and RSL3 on the viability, lipid peroxidation and apoptosis of DDLPS cell lines. The results revealed that erastin and RSL3 induced lipid peroxidation and apoptosis, thereby exerting cytotoxic effects. In addition, nutlin-3, an MDM2 inhibitor, was demonstrated to increase lipid peroxidation and cytotoxicity when applied prior to erastin treatment. Notably, nutlin-3 also upregulated SLC3A2 expression in DDLPS cell lines, thereby enhancing cystine uptake. This increase in cystine uptake was suppressed by erastin. In addition, nutlin-3-induced SLC3A2 upregulation was abolished by TP53 knockdown. Nutlin-3 combined with erastin or RSL3 reduced absolute p-4EBP-1 levels in NDDLS-1 cells and p-p70S6 levels in both cell lines, with no significant impact on the p-4EBP-1/4EBP-1 and p-p70S6/p70S6 ratios. These results indicate that ferroptosis is a therapeutic vulnerability in the response to MDM2 inhibition in DDLPS. Furthermore, combining MDM2 inhibitors with ferroptosis-inducing agents may provide a potential therapeutic strategy for DDLPS and the role of mTOR in the pro-apoptotic effect of these combinations deserve further investigation.
Collapse
Affiliation(s)
- Chueh-Chuan Yen
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Paul Chih-Hsueh Chen
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - San-Chi Chen
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Wen-Chi Wu
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Chiao-Han Yen
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Yung-Chan Lin
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Po-Kuei Wu
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Chao-Ming Chen
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Jir-You Wang
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Ta-Chung Chao
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Muh-Hwa Yang
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | | |
Collapse
|
13
|
Luo A, Zheng W, Zhang Q, Yuan Y, Li M, Du K, Gao A, Pei L, Xie J, Chen W, Zhang L, Guo X, Yang X, Zeng C, Yang G, Deng M. COPS5 Triggers Ferroptosis Defense by Stabilizing MK2 in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416360. [PMID: 40198582 PMCID: PMC12165036 DOI: 10.1002/advs.202416360] [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] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Sorafenib, which is proven to serve as a potent ferroptosis inducer, is used as a first-line treatment for patients with advanced hepatocellular carcinoma (HCC), but it has limited clinical benefits, mainly due to drug resistance. Herein, using genome-wide CRISPR/Cas9 knockout screening and multiple functional studies, this work identifies COP9 signalosome subunit 5 (COPS5) as a driver of sorafenib resistance and a suppressor of ferroptosis in HCC. Consistently, the amplification and overexpression of COPS5 are frequently observed in clinical HCC samples, which are associated with poor patient prognosis and might predict patient response to sorafenib therapy. Mechanistically, COPS5 stabilized mitogen-activated protein kinase 2 (MK2) through deubiquitination and, in turn, induced the activation of heat shock protein beta-1 (HSPB1), a ferroptosis repressor, thereby protecting HCC cells from ferroptosis and consequently leading to sorafenib resistance and tumor progression, while its own expression could be induced by sorafenib treatment via activating transcription factor 4 (ATF4)-activated transcription. Furthermore, pharmacological inhibition of COPS5/MK2 synergize with sorafenib to induce ferroptosis and suppress HCC progression. This data reveals the crucial role of COPS5 in triggering ferroptosis defense and sorafenib resistance through the activation of the MK2-HSPB1 axis in HCC and highlights the potential of targeting COPS5/MK2 combined with sorafenib as a promising strategy for treating HCC.
Collapse
Affiliation(s)
- Ai‐Ling Luo
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
- Department of Hematology and OncologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangdong Provincial Clinical Research Center for Child HealthGuangzhou510623China
| | - Wen‐Ying Zheng
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Qiong Zhang
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Yan Yuan
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Mei‐Qi Li
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Kai Du
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - An‐Ran Gao
- Department of Laboratory MedicineShunde HospitalGuangzhou University of Chinese MedicineFoshan528300China
| | - Li‐Jun Pei
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Jie Xie
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Wen‐Hao Chen
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Long Zhang
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Xiu‐Zhu Guo
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Xiao‐Ran Yang
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Chao Zeng
- Department of Pathologythe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Guo‐Hua Yang
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| | - Min Deng
- Guangzhou Institute of Cancer Researchthe Affiliated Cancer HospitalGuangzhou Medical UniversityGuangzhou510095China
| |
Collapse
|
14
|
Zheng Y, Li Q, Jin X, Zhu M, Liang Q, Wu Y, Pan F, Qiu H, Wang X, Lu D, Huang H. W-GA nanodots with multienzyme activities alleviate the inflammatory microenvironment in the treatment of acute wounds. Mater Today Bio 2025; 32:101662. [PMID: 40166380 PMCID: PMC11957797 DOI: 10.1016/j.mtbio.2025.101662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
Acute wounds present a significant clinical challenge due to delayed healing, which is often exacerbated by elevated levels of reactive oxygen species (ROS). These high ROS concentrations hinder the natural healing process, leading to prolonged recovery and increased risk of complications. W-GA nanodots, synthesized via a simple coordination method, have emerged as promising solutions, demonstrating multifunctional enzymatic activity that effectively scavenges ROS. To explore the underlying mechanisms of ROS-induced oxidative stress, we conducted RNA sequencing on macrophages exposed to H2O2. The results revealed significant regulation of key stress response pathways, including substantial upregulation of the "p53 signaling pathway" and the "HIF-1 signaling pathway," both of which are essential for cellular adaptation to oxidative stress. By alleviating oxidative stress, W-GA nanodots not only accelerate wound repair but also improve overall healing outcomes. Notably, RNA sequencing of animal tissue samples revealed that W-GA nanodots activate the "Wnt signaling pathway," further promoting wound healing. These findings underscore the potential of W-GA nanodots as a novel therapeutic strategy for enhancing wound healing and treating oxidative stress-related conditions, positioning them as promising candidates for future clinical applications in wound care and inflammatory diseases.
Collapse
Affiliation(s)
- Yang Zheng
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Qingrong Li
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, PR China
| | - Xu Jin
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Mengmei Zhu
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, PR China
| | - Qian Liang
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Yingjie Wu
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Fuqiang Pan
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Houhuang Qiu
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, PR China
| | - Decheng Lu
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Huiqiao Huang
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| |
Collapse
|
15
|
Li Y, Pan L, Zhao Q, Xiong H, Fang X, Guo X, Wang Y, Su X, Liu P, Hao J, Wang L, Wang X, Dong Q, Li Z, Jin F. Tanshinone IIA + Osthole alleviates ferroptosis in LPS-induced acute lung injury by Keap1-Nrf2/HO-1 pathway. Microb Pathog 2025; 203:107347. [PMID: 40147558 DOI: 10.1016/j.micpath.2025.107347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Acute lung injury (ALI) is associated with a high mortality rate and requires effective treatment. Tanshinone IIA (T) and Osthole (O) exhibit anti-inflammatory effects and have been used to protect against lipopolysaccharide (LPS)-induced lung injury in mice. However, the combined effects of T and O on lung injury protection and their potential protective mechanisms have not been studied. OBJECTIVE To assess the protective effects of TO on LPS-induced ALI in mice and BEAS-2B cell injury and to investigate the potential mechanisms underlying these protective effects. METHODS Models of ALI induced by LPS were established. The assessment encompassed the viability of BEAS-2B cells, cell count, myeloperoxidase (MPO) activity, protein content, as well as IL-6 and TNF-a levels in bronchoalveolar lavage fluid (BALF). Additionally, malondialdehyde (MDA), reactive oxygen species (ROS), and glutathione (GSH) levels in mouse lung tissue were measured. The effects of TO were assessed using immunofluorescence (IF), immunohistochemistry (IHC), Western Blot (WB), RT-PCR, and ELISA. Statistical analysis involved one-way ANOVA and t-test. RESULTS TO administration led to a significant reduction in lung edema (W/D), MDA, ROS, GSH, and superoxide dismutase (SOD) levels compared to the individual T or O groups, alleviating LPS-induced ALI. TO also significantly attenuated lung tissue damage, reduced inflammatory response, decreased Fe2+ and 4-HNE levels, and increased GPX4, SLC7A11, and Nrf2 gene expression in mice. Ultimately, TO alleviated ferroptosis in LPS-induced ALI by activating Nrf2 expression, and no markedly adverse reactions were observed. CONCLUSION TO alleviates LPS-induced ALI and effectively treats against LPS-induced ALI.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Lei Pan
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Qi Zhao
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Huanqing Xiong
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xiaoan Fang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xian Guo
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Ying Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xiaorong Su
- Department of Endocrinology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Pan Liu
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Jing Hao
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Lei Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xinqun Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Quandi Dong
- General Surgery Department, 947 Army Hospital, China.
| | - Zhichao Li
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China.
| | - Faguang Jin
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China.
| |
Collapse
|
16
|
Li BR, Wang T, Hu HF, Wu D, Zhou CJ, Ji SR, Zhuo QF, Li Z, Wang ZL, Fan GX, Jing DS, Yu CY, Qin Y, Chen XM, Xu JF, Xu XW. Acyl-CoA thioesterase 8 induces gemcitabine resistance via regulation of lipid metabolism and antiferroptotic activity in pancreatic ductal adenocarcinoma. Acta Pharmacol Sin 2025; 46:1742-1756. [PMID: 39939803 PMCID: PMC12098905 DOI: 10.1038/s41401-025-01477-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/08/2025] [Indexed: 02/14/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) comprises a group of highly malignant tumors of the pancreas. Metabolic reprogramming in tumors plays a pivotal role in promoting cancer progression. However, little is known about the metabolic alterations in tumors that drive cancer drug resistance in patients with PDAC. Here, we identified acyl-CoA thioesterase 8 (ACOT8) as a key player in driving PDAC gemcitabine (GEM) resistance. The expression of ACOT8 is significantly upregulated in GEM-resistant PDAC tissues and is closely associated with poor survival in patients with PDAC. Gain- and loss-of-function studies have shown that ACOT8 drives PDAC GEM resistance both in vitro and in vivo. Mechanistically, ACOT8 regulates cellular cholesterol ester (CE) levels, decreases the levels of phosphatidylethanolamines (PEs) that bind to polyunsaturated fatty acids and promote peroxisome activation. The knockdown of ACOT8 promotes ferroptosis and increases the chemosensitivity of tumors to GEM by inducing ferroptosis-associated pathway activation in PDAC cell lines. The combination of orlistat, an ACOT8 inhibitor, and GEM significantly inhibited tumor growth in PDAC organoid and mouse models. This study reveals the biological importance of ACOT8 and provides a potential combination therapy for treating patients with advanced GEM-resistant PDAC.
Collapse
Affiliation(s)
- Bo-Rui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Hepatopancreatobiliary Surgery, First College of Clinical Medical Science, Three Gorges University, Yichang, 443003, China
- People's Hospital of China Three Gorges University, Yichang, 443099, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Ting Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Hai-Feng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
- Department of General Surgery, First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Di Wu
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Chen-Jie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Shun-Rong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Qi-Feng Zhuo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zhi-Liang Wang
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Gui-Xiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - De-Sheng Jing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chong-Yuan Yu
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xue-Min Chen
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China.
| | - Jun-Feng Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xiao-Wu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Chen Y, Wu Y, Dong J, Zhang C, Tang J. Acacetin Attenuates Cigarette Smoke Extract-Induced Human Bronchial Epithelial Cell Injury by Activating NRF2/SLC7A11/GPX4 Signaling to Inhibit Ferroptosis. Cell Biochem Biophys 2025; 83:2499-2510. [PMID: 39751740 DOI: 10.1007/s12013-024-01659-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) stands as a major contributor to mortality worldwide, with cigarette smoke being a primary causative factor. Acacetin has been reported to possess lung protective effects. However, the precise role and mechanism of Acacetin in COPD remains elusive. In this study, human bronchial epithelial cell line HBE135-E6E7 was treated with Acacetin under cigarette smoke extract (CSE) conditions. Cellular viability was assessed using CCK-8 and LDH kits. Reactive oxygen species (ROS) generation was tested with DCFH-DA staining. JC-1 staining was employed to examine the mitochondrial membrane potential (MMP). Additionally, hydroxynonenal (4-HNE) level was tested using immunofluorescence staining and mitochondrial lipid peroxidation was evaluated using MitoPeDPP staining. MitoSOX staining was used to detect mitochondrial (mito)-ROS. Fe2+ level was measured using FerroOrange staining and the expression of ferroptosis-related proteins was detected with western blot. Besides, the binding between Acacetin and NRF2 was analyzed by molecular docking. The sequent NRF2 overexpression or knockdown was used to explore the regulation of Acacetin on NRF2/SLC7A11/GPX4 signaling. Results indicated that CSE significantly reduced the viability, augmented ROS generation and decreased MMP in HBE135-E6E7 cells, which were blocked by Acacetin addition. Moreover, Acacetin inhibited lipid peroxidation and ferroptosis in CSE-treated HBE135-E6E7 cells. Specifically, Acacetin targeted NRF2 and activated the NRF2/SLC7A11/GPX4 signaling in CSE-induced HBE135-E6E7 cells. Furthermore, NRF2 deficiency or ML-385 treatment notably restored the influences of Acacetin on oxidative stress and ferroptosis in HBE135-E6E7 cells challenged with CSE. In conclusion, Acacetin alleviated CSE-induced injury in HBE135-E6E7 cells by activating The NRF2/SLC7A11/GPX4 signaling to inhibit ferroptosis.
Collapse
Affiliation(s)
- Yongchang Chen
- Department of Pulmonary Disease, Yangzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, 225000, Jiangsu, China
| | - Yan Wu
- Department of Pulmonary Disease, Yangzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, 225000, Jiangsu, China
| | - Juan Dong
- Department of Traditional Chinese Medicine Culture Publicity, Yangzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, 225000, Jiangsu, China
| | - Chuanming Zhang
- Department of Pulmonary Disease, Yangzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, 225000, Jiangsu, China
| | - Jia Tang
- Yangzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, 225000, Jiangsu, China.
| |
Collapse
|
18
|
Gu D, Sun Y, Wang J, Sun J, Lou H, Kang W. Metformin regulates ferroptosis in Skin cutaneous melanoma via ATF3/NRF2 axis. Cancer Genet 2025; 294-295:136-144. [PMID: 40318300 DOI: 10.1016/j.cancergen.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND To explore the effects of metformin on the proliferation and ferroptosis of skin cutaneous melanoma (SKCM) and its potential molecular mechanisms, providing a new theoretical basis and strategy for the treatment of cutaneous melanoma. METHODS The CCK-8 experiment was used to detect the effect of metformin on the proliferation of skin cutaneous melanoma cells. Kits were used to detect glutathione (GSH) content, reactive oxygen species (ROS), lipid peroxide (LPO), and malondialdehyde (MDA) levels to evaluate ferroptosis-related indicators. RNA-seq sequencing and related analyses were used to screen differentially expressed genes and explore their involved biological functions and signaling pathways. Western blot was used to detect the expression levels of ATF3 and NRF2 proteins and analyze the regulatory effect of metformin on the ATF3/NRF2 axis. RESULTS Metformin significantly reduced the proliferation ability of skin cutaneous melanoma cells. The treated cells showed a decrease in GSH content and an accumulation of ROS, LPO, and MDA, suggesting that ferroptosis was regulated. RNA-seq analysis found 2068 differentially expressed genes, of which 897 were up-regulated and 1171 were down-regulated. The related pathways such as iron metabolism disorders and ferroptosis were activated. After metformin treatment, the expression of ATF3 mRNA in cells increased and was positively correlated with the concentration, while the expression in SKCM tissues decreased. At the same time, the expression of ATF3 protein increased and the expression of NRF2 protein decreased, suggesting that metformin may induce ferroptosis through the ATF3/NRF2 axis. CONCLUSION Metformin can induce ferroptosis by regulating ATF3/NRF2 axis, which may be a novel strategy for improving the treatment of skin cutaneous melanoma.
Collapse
Affiliation(s)
- Da Gu
- Department of Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, PR China
| | - Yulin Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Jianghui Wang
- Department of Orthopedic Surgery, The Second People's Hospital of Dongying, Dongying, Shandong 257000, PR China
| | - Jinpeng Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Huanmin Lou
- Department of Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, PR China.
| | - Weiting Kang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China.
| |
Collapse
|
19
|
He Z, Long H, Ma H, Ran W, Song Y, Shang W, Wang J, Qin Y, Deng Z. 4, 9-dihydroxy-α-lapachone as a potent antiproliferation agent for triple-negative breast cancer via ferroptosis. Fitoterapia 2025; 183:106567. [PMID: 40280251 DOI: 10.1016/j.fitote.2025.106567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant breast cancer. Ferroptosis is an oxidative, iron-dependent form of regulated cell death. Ferroptosis-targeted therapies is a promising approach to improving treatment outcomes of TNBC. Combining death pathway inhibitors with relevant indices for ferroptosis and LipROS, this study uncovered that a natural product of 4, 9-dihydroxy-α-lapachone (DLN) from Catalpa bungei "jinsi" exhibited in vitro and in vivo inhibitory activity against TNBC via ferroptosis. The molecular mechanism is an activation of the FTH1 led to iron overload, and then inhibition of cysteine-glutamate antiporter (system Xc-) and GPX4, which further sensitized TNBC cells to ferroptosis. This study clarified the pathway of DLN-induced cell death in TNBC treatment and exhibited its potential as therapeutic agent for TNBC.
Collapse
Affiliation(s)
- Zhong He
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hui Long
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Hongyue Ma
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Wanrong Ran
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yinhong Song
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, China.
| | - Wenjuan Shang
- Medical College, Hubei Three Gorges Polytechnic, Yichang 443000, China
| | - Junhui Wang
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation of State Forestry Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing 100091, China.
| | - Ye Qin
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Zhangshuang Deng
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
20
|
Zhao H, Wang Z, Wang H. The role of NCOA4-mediated ferritinophagy in the ferroptosis of hepatocytes: A mechanistic viewpoint. Pathol Res Pract 2025; 270:155996. [PMID: 40344841 DOI: 10.1016/j.prp.2025.155996] [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: 02/07/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
This paper focuses on the mechanism underlying nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and subsequent hepatocyte ferroptosis. Iron is a pivotal trace element, but excessive iron deposition can lead to liver injury. Ferroptosis is a recognized, iron-dependent mode of programmed cell death that plays an important role in various liver diseases. NCOA4 is a key molecule mediating the selective autophagic degradation of ferritin. It affects ferroptosis by regulating intracellular free iron levels. NCOA4 expression is regulated by various factors, including cellular iron levels and oxidative stress. It was demonstrated that inhibition of NCOA4 can reduce iron-mediated cell death and mitigate liver damage, suggesting that NCOA4 may be a potential target for the prevention and treatment of liver diseases. Further in-depth studies of the molecular mechanism of NCOA4-mediated ferritinophagy and its relationship with iron-induced cell death can provide novel ideas for the diagnosis and treatment of liver diseases. The deficiency or abnormal expression of NCOA4 is closely associated with ferroptosis in a variety of liver diseases, including non-alcoholic fatty liver disease, alcoholic liver disease, drug-induced liver injury, and liver fibrosis. Future studies should focus on elucidating the dynamic changes in the NCOA4 regulatory network during specific pathological processes. This strategy can lay the foundation for drug development.
Collapse
Affiliation(s)
- Huixian Zhao
- Department of Hepatopancreatobiliary Surgery, the Affiliated Hospital of Qinghai University, China; Qinghai Research Key Laboratory for Echinococcosis, China; Qinghai Province Women and Children's hospital, China
| | - Zhixin Wang
- Department of Hepatopancreatobiliary Surgery, the Affiliated Hospital of Qinghai University, China; Qinghai Research Key Laboratory for Echinococcosis, China.
| | - Haijiu Wang
- Department of Hepatopancreatobiliary Surgery, the Affiliated Hospital of Qinghai University, China; Qinghai Research Key Laboratory for Echinococcosis, China.
| |
Collapse
|
21
|
Zhang H, Wang Y, Jiang M, Wang K, Yan J, Li G, Zheng Z. Inherently anti-metastatic peptide hydrogels for sonodynamic-amplified ferroptosis in cancer therapy. Mater Today Bio 2025; 32:101688. [PMID: 40206142 PMCID: PMC11980000 DOI: 10.1016/j.mtbio.2025.101688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer metastasis remains a significant challenge in oncology, prompting the exploration of innovative biomaterials to enhance treatment efficacy. While many hydrogels only serve as passive carriers, this study presents two novel self-assembling peptides, CWEWTWY and NapFFSGP, which form supramolecular hydrogels with intrinsic anti-metastatic properties. We demonstrate a correlation between the nanofibrous morphology of these peptides and their enhanced anti-metastatic activity, mediated by disruption of F-actin organization and impacting pathways related to cancer cell adhesion and actin filament dynamics. In vivo studies confirm a significant reduction in lung metastasis using a 4T1 pulmonary metastasis model. We also demonstrate their potential as a simple, synergistic platform integrating sonodynamic therapy (SDT) and ferroptosis. Ironporphyrin (FP), incorporated into Gel@FP, acts as both a sonosensitizer and ferroptosis inducer. Upon ultrasound irradiation, FP generates localized reactive oxygen species, further amplifying ferroptosis through enhanced lipid peroxidation. Gel@FP combined with ultrasound demonstrates potent antitumor efficacy in vitro and in vivo, promoting apoptosis, ferroptosis, and immunogenic cell death, leading to enhanced tumor regression and robust immune activation. Our findings highlight the potential of anti-metastatic hydrogels as a promising multifunctional platform to address the challenges of metastasis while enhancing antitumor immunity.
Collapse
Affiliation(s)
- Hongxia Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yamei Wang
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Mengmeng Jiang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Kunyu Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Jingru Yan
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Gongyu Li
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhen Zheng
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
22
|
Zeng W, Wang F, Cui Z, Zhang Y, Li Y, Li N, Mao Z, Zhang H, Liu Y, Miao Y, Sun S, Cai Y, Xiong B. Inhibition of ferroptosis counteracts the advanced maternal age-induced oocyte deterioration. Cell Death Differ 2025; 32:1071-1085. [PMID: 39910323 PMCID: PMC12162888 DOI: 10.1038/s41418-025-01456-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
Ferroptosis, a recently discovered form of programmed cell death triggered by the excessive accumulation of iron-dependent lipid peroxidation products, plays a critical role in the development of various diseases. However, whether it is involved in the age-related decline in oocyte quality remains unexplored. Here, we took advantage of nano-proteomics to uncover that reduced ferritin heavy chain (Fth1) level is a major cause leading to the occurrence of ferroptosis in aged oocytes. Specifically, induction of ferroptosis in young oocytes by its activators RSL3 and FAC, or knockdown of Fth1 all phenocopied the meiotic defects observed in aged oocytes, including failed oocyte meiotic maturation, aberrant cytoskeleton dynamics, as well as impaired mitochondrial function. Transcriptome analysis showed that knockdown of Fth1 affected meiosis-related and aging-related pathways in oocytes. Conversely, inhibition of ferroptosis by its inhibitors or expression of Fth1 improved the quality of aged oocytes. We also validated the effects of ferroptosis on the porcine oocyte quality in vitro. Altogether, we demonstrate the contribution of ferroptosis to the age-induced oocyte defects and evidence that inhibition of ferroptosis might be a feasible strategy to ameliorate the reproductive outcomes of female animals at an advanced age.
Collapse
Affiliation(s)
- Wenjun Zeng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Feixue Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhaokang Cui
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yu Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Na Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zipeng Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Hanwen Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yiting Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yilong Miao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shaochen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yafei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
- College of Animal Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
23
|
Jeong J, Thi Quynh Mai N, Moon BS, Choi JK. Impact of polystyrene microplastics (PS-MPs) on the entire female mouse reproductive cycle: Assessing reproductive toxicity of microplastics through in vitro follicle culture. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118228. [PMID: 40315747 DOI: 10.1016/j.ecoenv.2025.118228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/05/2025] [Accepted: 04/20/2025] [Indexed: 05/04/2025]
Abstract
This study aims to investigate the effects of polystyrene microplastics (PS-MPs) on the entire female reproductive cycle and to elucidate the molecular mechanisms underlying their adverse impact on female ovaries. Additionally, it develops an in vitro follicle culture system as a novel methodological approach to evaluate reproductive toxicity, mimicking in vivo reproductive outcomes. First, PS-MPs were characterized using FTIR spectroscopy, TEM, and fluorescence microscopy. To assess reproductive toxicity, female mice were exposed to polystyrene microplastics (PS-MPs) at a dose of 30 mg/kg with an average particle size of 1 μm for 35 days. As a result, PS-MPs accumulated in the ovaries, leading to increased follicular atresia and apoptosis of granulosa cells. TEM revealed abnormal mitochondrial morphology in granulosa cells. Post-superovulation treatment, significant differences were noted in the number of ovulated metaphase II (MII) oocytes, spindle chromosome integrity, mitochondrial patterns, and ROS levels compared to controls. Mating with PS-MPs-exposed females led to fewer offspring. The in vitro follicle culture system proved promising for assessing PS-MPs reproductive toxicity. Immunohistochemistry showed increased Cleaved Caspase 3 and decreased Bcl2 levels in PS-MPs-treated groups, indicating apoptosis in granulosa cells. PS-MPs activate JNK and ERK pathways to mediate cell death, while impairing AKT signaling, reducing granulosa cell survival and ovarian function. This study highlights PS-MPs adverse reproductive effects and aids in developing strategies to protect female reproductive health.
Collapse
Affiliation(s)
- Jukyeong Jeong
- Department of Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Nhu Thi Quynh Mai
- Department of Medical Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Byoung-San Moon
- Department of Medical Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Jung Kyu Choi
- Department of Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
24
|
Meng Y, Zhou Q, Dian Y, Zeng F, Deng G, Chen X. Ferroptosis: A Targetable Vulnerability for Melanoma Treatment. J Invest Dermatol 2025; 145:1323-1344. [PMID: 39797894 DOI: 10.1016/j.jid.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 01/13/2025]
Abstract
Melanoma is a devastating form of skin cancer characterized by a high mutational burden, limited treatment success, and dismal prognosis. Although immunotherapy and targeted therapies have significantly revolutionized melanoma treatment, the majority of patients fail to achieve durable responses, highlighting the urgent need for novel therapeutic strategies. Ferroptosis, an iron-dependent form of regulated cell death driven by the overwhelming accumulation of lipid peroxides, has emerged as a promising therapeutic approach in preclinical melanoma models. A deeper understanding of the ferroptosis landscape in melanoma based on its biology characteristics, including phenotypic plasticity, metabolic state, genomic alterations, and epigenetic changes, as well as the complex role and mechanisms of ferroptosis in immune cells could provide a foundation for developing effective treatments. In this review, we outline the molecular mechanisms of ferroptosis, decipher the role of melanoma biology in ferroptosis regulation, reveal the therapeutic potential of ferroptosis in melanoma, and discuss the pressing questions that should guide future investigations into ferroptosis in melanoma.
Collapse
Affiliation(s)
- Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| |
Collapse
|
25
|
Yang H, Yuan D, Zhou Z, Zhao H. Nitrate enrichment exacerbates microbiome and metabolism disturbances of the coral holobiont under heat stress. MARINE ENVIRONMENTAL RESEARCH 2025; 208:107098. [PMID: 40139064 DOI: 10.1016/j.marenvres.2025.107098] [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: 01/19/2025] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Coral reef ecosystems are facing severe deterioration due to escalating global temperatures and human-induced activities. Combined nitrate and heat stress can exacerbate coral bleaching, however, the underlying mechanism is still unclear. In the present study, we assessed the bleaching status of Acropora hyacinthus, a reef-building coral species, under high temperature and nitrate stress conditions using chemostat cultivation. We observed nitrate enrichment (9 μM) induced a significant reduction in photosystem efficiency (Fv/Fm) of Symbiodiniaceae and an increased thermal bleaching of corals under high temperature (30 °C). Nitrate exposure promoted the proliferation of Enterobacteriaceae and Vibrionaceae, which are bacterial families, potentially augmenting the coral's susceptibility to disease while exerting negligible effects on the fungal community. Alterations were observed in the metabolic pathways of both the coral hosts and Symbiodiniaceae, including down-regulated folate biosynthesis and inflammatory mediator regulation of TRP channels. Our findings indicate that nitrate enrichment under heat stress disrupts the metabolism of coral holobionts through altering bacterial communities, ultimately leading to increased coral bleaching.
Collapse
Affiliation(s)
- Huidan Yang
- Hainan International Joint Research Center for Coral Reef Ecology, Hainan University, Haikou, 570228, China; Center for Eco-Environment Restoration of Hainan Province, School of Ecology, Hainan University, Haikou, 570228, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China
| | - Dongdan Yuan
- Hainan International Joint Research Center for Coral Reef Ecology, Hainan University, Haikou, 570228, China; Center for Eco-Environment Restoration of Hainan Province, School of Ecology, Hainan University, Haikou, 570228, China
| | - Zhuojing Zhou
- Hainan International Joint Research Center for Coral Reef Ecology, Hainan University, Haikou, 570228, China; Center for Eco-Environment Restoration of Hainan Province, School of Ecology, Hainan University, Haikou, 570228, China
| | - Hongwei Zhao
- Hainan International Joint Research Center for Coral Reef Ecology, Hainan University, Haikou, 570228, China; Center for Eco-Environment Restoration of Hainan Province, School of Ecology, Hainan University, Haikou, 570228, China.
| |
Collapse
|
26
|
Wang ZY, Zeng T, Wang X, Zhuo X, Zheng JW, Zhu L, Cheng ST, Wan LH. Targeting inducible nitric oxide synthase with 1400W mitigates septic acute lung injury through inhibiting SLC7A11/GPX4 mediated ferroptosis. Nitric Oxide 2025; 158:4-10. [PMID: 40447183 DOI: 10.1016/j.niox.2025.05.008] [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: 04/23/2025] [Revised: 05/21/2025] [Accepted: 05/27/2025] [Indexed: 06/02/2025]
Abstract
Accumulating evidence has indicated that lung ferroptosis is an important contributor to septic acute lung injury (SALI). Inducible nitric oxide synthase (iNOS) may be implicated in the regulation of bronchial epithelial ferroptosis. Nevertheless, the precise mechanisms by which iNOS modulates ferroptosis remain elusive. This study investigated whether iNOS selective inhibitor 1400w alleviates LPS-induced SALI and suppresses ferroptosis in mice. Additionally, RNA sequencing (RNA-seq), molecular docking, molecular dynamic simulation, Transmission electron microscope (TEM), and western blotting were employed to predict and evaluate the molecular mechanism of 1400w on LPS-induced ferroptosis in vivo. The results showed that the administration of 1400w markedly attenuated LPS-induced lung injury and facilitated pulmonary function in mice. Also, 1400w administration effectively suppressed bronchial epithelial ferroptosis induced by LPS in mice. Furthermore, molecular docking and molecular dynamics simulations revealed stable binding between GPX4 and iNOS, with 1400w modulating ferroptosis mediated by SLC7A11/GPX4 through targeting iNOS. Collectively, our research demonstrated that inhibition of iNOS might represent a potential therapeutic strategy to improve SALI by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Zi-Yao Wang
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China; NHC Key Laboratory of Chronobiology (Sichuan University), West China School of Basic Medical Sciences & Forensic Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Tao Zeng
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xin Wang
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xin Zhuo
- NHC Key Laboratory of Chronobiology (Sichuan University), West China School of Basic Medical Sciences & Forensic Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Jing-Wen Zheng
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China; NHC Key Laboratory of Chronobiology (Sichuan University), West China School of Basic Medical Sciences & Forensic Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Ling Zhu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Shu-Ting Cheng
- NHC Key Laboratory of Chronobiology (Sichuan University), West China School of Basic Medical Sciences & Forensic Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China.
| | - Li-Hong Wan
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China; NHC Key Laboratory of Chronobiology (Sichuan University), West China School of Basic Medical Sciences & Forensic Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China.
| |
Collapse
|
27
|
Liao T, Xu X, Wang G, Yan J. p53-mediated suppression of the SLC7 A11/GPX4 signaling pathway promotes trophoblast ferroptosis in preeclampsia. BMC Biol 2025; 23:141. [PMID: 40437575 PMCID: PMC12121027 DOI: 10.1186/s12915-025-02240-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/09/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Ferroptosis is an iron-dependent form of non-apoptotic cell death that occurs through increased plasma membrane phospholipid peroxidation in the context of impaired plasma membrane phospholipid peroxide repair systems. It has been reported that p53 can inhibit the expression of cysteine/glutamate reverse transporter solute carrier family 7, member 11 (SLC7A11), a key component of system Xc-, thus inhibiting cysteine uptake and promoting reactive oxygen species (ROS) accumulation as an important part of cell ferroptosis. Preeclampsia (PE) is an idiopathic hypertensive disease of pregnancy. Spiral artery insufficiency and impaired placental development are present at all stages, leading to placental hypoperfusion, ischemia, and hypoxia. However, the role of ferroptosis, particularly p53-mediated trophoblast ferroptosis, in placental dysfunction during PE remains unclear. RESULTS In PE placental tissues, malondialdehyde (MDA) and total iron levels were elevated, and trophoblasts exhibited typical ferroptosis-associated morphological changes. Additionally, p53 mRNA and protein expression and the percentage of p53-positive cells were increased, while SLC7A11 and GPX4 mRNA and protein expression and the percentage of positive cells were decreased. VEGFR1 protein expression was upregulated, whereas VEGFA and PLGF protein expression was downregulated. p53 protein expression was negatively correlated with the expression of proteins in the SLC7A11/GPX4 signaling pathway, VEGFA, and PLGF. Conversely, there was a positive correlation between p53 expression and MDA, total iron concentration, and VEGFR1. In vitro, the ferroptosis inducer erastin increased ROS levels in trophoblast cells. The ferroptosis inhibitor Fer-1, the apoptosis inhibitor Z-VAD-FMK, and the necrosis inhibitor Nec-1 failed to prevent erastin-induced ROS elevation. In p53 + / + trophoblasts, erastin-induced ROS elevation was more pronounced than that in p53 - / - and control cells, and angiogenesis was impaired. In pregnant rats, p53 + / + placentas exhibited increased MDA and total iron levels, ferroptosis-like morphological changes in trophoblasts, and reduced CD34 expression. p53 protein expression was negatively correlated with CD34 expression. CONCLUSION This study confirmed that trophoblast ferroptosis occurs in the pathological state of PE and that trophoblast are specifically sensitive to ferroptosis. p53 can mediate the SLC7A11/GPX4 signaling pathway to promote ferroptosis of trophoblast cells in the pathogenesis of PE. It is also speculated that increased p53 reactivity may mediate impaired angiogenesis in placental tissues.
Collapse
Affiliation(s)
- Tingting Liao
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University Fujian Maternity and Child Health Hospital, Fujian Clinical Research Center for Maternal-Fetal Medicine, Laboratory of Maternal-Fetal Medicine, Fujian Maternity and Child Health Hospital, National Key Obstetric Clinical Specialty Construction Institution of China, No. 18, Daoshan Road, Gulou District, Fuzhou, Fujian Province, China
| | - Xia Xu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University Fujian Maternity and Child Health Hospital, Fujian Clinical Research Center for Maternal-Fetal Medicine, Laboratory of Maternal-Fetal Medicine, Fujian Maternity and Child Health Hospital, National Key Obstetric Clinical Specialty Construction Institution of China, No. 18, Daoshan Road, Gulou District, Fuzhou, Fujian Province, China
| | - Guiying Wang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University Fujian Maternity and Child Health Hospital, Fujian Clinical Research Center for Maternal-Fetal Medicine, Laboratory of Maternal-Fetal Medicine, Fujian Maternity and Child Health Hospital, National Key Obstetric Clinical Specialty Construction Institution of China, No. 18, Daoshan Road, Gulou District, Fuzhou, Fujian Province, China
| | - Jianying Yan
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University Fujian Maternity and Child Health Hospital, Fujian Clinical Research Center for Maternal-Fetal Medicine, Laboratory of Maternal-Fetal Medicine, Fujian Maternity and Child Health Hospital, National Key Obstetric Clinical Specialty Construction Institution of China, No. 18, Daoshan Road, Gulou District, Fuzhou, Fujian Province, China.
| |
Collapse
|
28
|
He W, Hu X, Ge M, Meng K. The central role of ferroptosis-induced therapy mediated by tenacissoside H in anaplastic thyroid cancer. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119908. [PMID: 40311717 DOI: 10.1016/j.jep.2025.119908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/20/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tenacissoside H(TDH), a natural compound extracted from the dried vine stems of Marsdenia tenacissima (Roxb.) Wight et Arn., is considered to have anti-tumor effects. However, the anti-tumor activity of TDH against ATC remains unknown. AIM OF THE STUDY Ferroptosis, a novel form of programmed cell death, presents a promising target for therapeutic intervention, particularly in overcoming drug resistance in anaplastic thyroid carcinoma (ATC). We investigated the inhibitory effects of TDH on ATC cells, elucidating its ferroptosis-inducing mechanism, which to our knowledge has not been explored before. RESULTS Our findings indicate that TDH exerts an effect on the survival, proliferation, and migration of ATC cells. The strength of effect is dependent on dosage. Notably, ferroptosis marker proteins (GPX4, xCT, HO-1, TFR) were significantly downregulated following TDH treatment, whereas GPX4 and xCT expressions were partially restored post treatment with ferrostatin-1. Furthermore, in vivo studies confirmed that TDH effectively inhibited tumor growth in xenografted 8505C cells. CONCLUSIONS TDH could be considered a potential agent against ATC via inducing ferroptosis, providing a novel pharmacological basis for treating ATC.
Collapse
Affiliation(s)
- Wanwan He
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Xiaotian Hu
- Shanghai First People's Hospital, Shanghai, 200080, China
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China.
| | - Kexin Meng
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China.
| |
Collapse
|
29
|
Patil N, Bhatt LK. Targeting Acyl-CoA synthetase long-chain family member 4: a potential approach for the treatment of cerebral ischemia/reperfusion injury. Metab Brain Dis 2025; 40:212. [PMID: 40418418 DOI: 10.1007/s11011-025-01638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
Cerebral ischemia/reperfusion injury causes high rates of morbidity and death. Recent studies have shown that ferroptosis, a type of controlled cell death brought on by lipid peroxidation, worsens cerebral ischemia/reperfusion injury. Acyl-CoA synthetase long-chain family member 4 (ACSL4) has emerged as a crucial enzyme in lipid metabolism and ferroptosis in the context of ischemia/reperfusion injury, influencing neuronal cell death. Increased vulnerability to ferroptosis and worsening ischemia/reperfusion injury outcomes are linked to elevated ACSL4 levels. Comprehending the molecular processes underlying ACSL4-mediated ferroptosis may result in novel approaches to treating cerebral ischemia/reperfusion injury. The present review discusses ACSL4 as a potential target for treating cerebral ischemia/reperfusion injury, focusing on ACSL4-mediated ferroptosis and signal transduction.
Collapse
Affiliation(s)
- Nikita Patil
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
30
|
Xu X, Zhang H, Chang A, Peng H, Li S, Zhang K, Wang W, Yin X, Qu C, Dong X, Ni J. Astilbin Alleviates IL-17-Induced Hyperproliferation and Inflammation in HaCaT Cells via Inhibiting Ferroptosis Through the cGAS-STING Pathway. Int J Mol Sci 2025; 26:5075. [PMID: 40507886 PMCID: PMC12155464 DOI: 10.3390/ijms26115075] [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: 04/22/2025] [Revised: 05/14/2025] [Accepted: 05/22/2025] [Indexed: 06/16/2025] Open
Abstract
Psoriasis, a chronic inflammatory skin disorder, is driven by dysregulated immune responses and keratinocyte dysfunction. Here, we explore the therapeutic potential of Astilbin (AST), a flavonoid with potent anti-inflammatory properties, in modulating ferroptosis and the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway in IL-17-stimulated HaCaT keratinocytes. Our psoriatic cell model recapitulated key pathological features, including hyperproliferation, membrane integrity loss, mitochondrial dysfunction, and heightened oxidative stress, alongside elevated proinflammatory cytokine levels. Ferroptosis-related biomarkers were significantly altered, with increased malondialdehyde (MDA) accumulation, reduced glutathione (GSH) levels, iron overload (Fe2+), and enhanced lipid peroxidation (detected via C11-BODIPY). Mechanistically, mitochondrial damage triggered cytoplasmic leakage of mitochondrial DNA (mtDNA), activating the cGAS-STING pathway, as evidenced by upregulated pathway-associated protein expression. AST intervention effectively mitigated these pathological changes by suppressing ferroptosis and modulating cGAS-STING signaling. These findings reveal a dual-pathway regulatory mechanism, positioning AST as a promising therapeutic candidate for psoriasis. By elucidating the interplay between ferroptosis and the cGAS-STING pathway, this study provides new insights into psoriatic inflammation and offers a rationale for targeting these pathways in therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xiaoxv Dong
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (X.X.); (H.Z.); (A.C.); (H.P.); (S.L.); (K.Z.); (W.W.); (X.Y.); (C.Q.)
| | - Jian Ni
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (X.X.); (H.Z.); (A.C.); (H.P.); (S.L.); (K.Z.); (W.W.); (X.Y.); (C.Q.)
| |
Collapse
|
31
|
Niu S, Guo M, Yang H, Chang X, Shang M, Wu L, Ma Y, Xue Y. Ferroptosis effects and behavioral changes associated with high accumulation and low elimination percentage of silver in brain following a 28-day nasal instillation of silver nanoparticles in mice. Food Chem Toxicol 2025; 202:115567. [PMID: 40409664 DOI: 10.1016/j.fct.2025.115567] [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: 03/09/2025] [Revised: 04/18/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
This study investigates the ferroptosis effects and behavioral changes in mice following 28-day nasal instillation of silver nanoparticles (AgNPs, 0-50 mg/kg.bw), with subsequent observation of recovery effects over a 28-day period. AgNP accumulates differentially in the organs evaluated. However, the hippocampus was an important target. Spatial learning and memory deficits were detected using the Morris Water Maze. Hippocampal neuronal damage was observed, characterized by loss of cellular integrity, cytoplasmic atrophy, and blurred nuclei. These alterations persisted throughout the 28-day recovery period. Lipid peroxidation was triggered by depletion of GSH and elevation of MDA. Disruption of iron homeostasis was mediated through downregulation of TFRC, FTH1, and FTL protein expression. Ferroptosis activation was confirmed by suppression of GPX4 and SLC7A11, coupled with upregulation of ACSL4 and COX2. Additionally, exposure to Ag+ in the amount released from AgNPs (50 mg/kg) also resulted in sustained silver accumulation and ferroptosis effects. Beyond nanoparticle-specific toxicity, the study emphasizes the potential adverse effects resulting from the prolonged and gradual release of metal ions from these nanoparticles in vivo. Our study provided experimental evidence and potential clues into the neurotoxicity of metal nanoparticles.
Collapse
Affiliation(s)
- Shuyan Niu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Menghao Guo
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Haitao Yang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xiaoru Chang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Mengting Shang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Liqing Wu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yu Ma
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
32
|
Yuan Q, Wang X, Xiao Y, Yang Z, Cai X, Gao W, Mi Y, Zhang C. Attenuating Effect of a Polyphenol Ellagic Acid on Ovarian Aging by Inhibiting the Ferroptosis Pathway in Low-Yield Laying Chickens. Antioxidants (Basel) 2025; 14:614. [PMID: 40427495 PMCID: PMC12109511 DOI: 10.3390/antiox14050614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/14/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025] Open
Abstract
Aging leads to ovarian degeneration in poultry, reducing egg production and quality. Ellagic acid (EA), a natural plant-derived compound, may help delay ovarian aging, though its precise mechanisms remain unclear. This study investigated the effects of EA on ovarian aging of low-yield laying chickens and explored its underlying mechanism. EA supplementation (100 and 500 mg/kg) significantly increased ovarian weight as well as the number and proportion of small yellow follicles in aging chickens. EA administration elevated serum antioxidant levels and upregulated the expression of glutathione peroxidase 4 (GPX4) expression to reduce oxidative stress. Importantly, EA treatment suppressed the mRNA and protein expression of ferroptosis markers transferrin receptor protein 1 (TFRC) and solute carrier family 7 member 11 (SLC7A11), increased Proliferating Cell Nuclear Antigen (PCNA) expression, and alleviated G1 phase arrest in granulosa cells (GCs), promoting cell proliferation, which improves egg quality and production. Furthermore, in vitro experiments demonstrated that EA treatment decreased reactive oxygen species production, improved mitochondrial function, inhibited ferroptosis, and attenuated GCs aging. In conclusion, this study reveals the critical role of ferroptosis in chicken ovarian aging and suggests that EA may provide a promising approach for delaying ovarian aging and enhancing productivity in low-yield poultry.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuling Mi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (Q.Y.); (X.W.); (Y.X.); (Z.Y.); (X.C.); (W.G.)
| | - Caiqiao Zhang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (Q.Y.); (X.W.); (Y.X.); (Z.Y.); (X.C.); (W.G.)
| |
Collapse
|
33
|
Xu J, Luo X, Su W, Jia G, Cai H, Li D, Li R, Wang X, Yang Y, Wang T, Zuo C. Turning Waste into Treasure: Radiation Byproduct-Induced Fe(III)/Fe(II) Conversion for Efficient Ferroptosis to Improve Iodine-131-Based Transarterial Radioembolization for Liver Tumors. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29170-29182. [PMID: 40338615 DOI: 10.1021/acsami.5c01445] [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: 05/09/2025]
Abstract
Transarterial radioembolization (TARE) is a primary palliative treatment for advanced liver cancer. Nonetheless, its therapeutic efficacy is frequently hindered by resistance to tumor cell apoptosis induced by inter-radiotherapy. Induction of multiple cell death modalities provides a potential solution to this challenge. Ferroptosis, a distinct form of cell death from apoptosis, is dependent on the intracellular Fe2+-mediated Fenton reaction for the production of hydroxyl radicals (·OH) and is gaining recognition as a promising approach for cancer treatment. In this study, we synthesized a therapeutic radionuclide iodine-131 (131I)-based TARE agent by combining 131I-labeled iron-based MIL-88B(Fe) nanoparticles (NPs) (abbreviated as 131I-MIL-88B(Fe)) with Lipiodol to achieve a combined apoptosis-ferroptosis tumor therapy. Specifically, a mixture of Lipiodol and 131I-MIL-88B(Fe) NPs was injected into the liver tumors through the hepatic artery. Lipiodol blocks the arterial blood supply of the tumor, causing tumor tissue necrosis, whereas 131I inter-radiotherapy damages deoxyribonucleic acid (DNA) through direct action or indirectly via the production of ·OH through H2O radiolysis, leading to tumor cell apoptosis. Importantly, hydrated electrons (eaq-), a byproduct of H2O radiolysis, promoted the conversion of Fe3+ to Fe2+ in MIL-88B(Fe) NPs, enhancing the efficacy of the Fenton reaction and triggering ferroptosis. In vitro experiments demonstrated that compared to 131I alone, 131I-MIL-88B(Fe) NPs significantly enhanced ferroptosis-mediated tumor cell death due to 131I-induced Fe2+ production, which increased catalytic activity in the Fenton reaction. In a rat model bearing orthotopic N1S1 liver tumors, TARE with Lipiodol and 131I-MIL-88B(Fe) NPs induced tumor cell necrosis, apoptosis, and ferroptosis, resulting in improved therapeutic outcomes. This study leverages eaq- to facilitate Fe3+/Fe2+ conversion for efficient ferroptosis, turning waste into a valuable resource. This demonstrated the innovative integration of multiple treatment strategies to augment the efficacy of TARE in liver cancer therapy.
Collapse
Affiliation(s)
- Jiangnan Xu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
- Department of Nuclear Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xiu Luo
- Department of Nuclear Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Weiwei Su
- Department of Nuclear Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Radiology, Naval Medical Centre, Shanghai 200052, China
| | - Guorong Jia
- Department of Nuclear Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Danni Li
- Department of Nuclear Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Rou Li
- Department of Nuclear Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xiangdong Wang
- Mini-Invasive Intervention Center, the Third Affiliated Hospital of Naval Medical University, Shanghai 200438, China
| | - Yefa Yang
- Mini-Invasive Intervention Center, the Third Affiliated Hospital of Naval Medical University, Shanghai 200438, China
| | - Tao Wang
- Department of Nuclear Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Changjing Zuo
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
- Department of Nuclear Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| |
Collapse
|
34
|
Pang Y, Ke Y, Amona FM, Chen X, Liu Z, Chen J, Liang Y, Wang F, Wang Y, Fang X, Chen X, Zhang C. Cyanidin-3-O-glucoside mitigates Staphylococcus aureus-induced mastitis by suppressing inflammatory responses and Ferroptosis mediated by SESN2/Nrf2. Int Immunopharmacol 2025; 159:114868. [PMID: 40394793 DOI: 10.1016/j.intimp.2025.114868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/25/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025]
Abstract
Mastitis is a significant concern in both human and animal medicine. The causative agent S. aureus is one of the most challenging pathogens responsible for mastitis, and the rise of its antibiotic resistance underscores the need for alternative therapies. The SESN2/Nrf2 pathway, owing to its pivotal role in regulating cellular antioxidant defenses, which are critically disrupted during ferroptosis, has recently received less attention. However, whether C3G targets the SESN2/Nrf2 pathway remains unclear, which provides a dual mechanism for treating S. aureus-induced mastitis by reducing inflammation and safeguarding mammary epithelial cells (MECs) from ferroptosis. Using a mouse mastitis and MECs model, we investigated the therapeutic potential of C3G in alleviating S. aureus-induced mastitis, focusing specifically on its role in inhibiting inflammation and modulating ferroptosis through the SESN2/Nrf2 pathway. The results demonstrated the potential antimicrobial effects of C3G against S. aureus and MRSA, suppressed inflammatory responses by downregulating pro-inflammatory markers (IL-1β, IL-6, and TNF-α), and inhibited STAT2/STAT3 signaling. Furthermore, C3G modulates ferroptosis by activating the SESN2/Nrf2 pathway, reducing oxidative stress, and protecting mammary epithelial cells from ferroptosis-induced damage. This comprehensive approach highlights C3G's potential as a novel therapeutic strategy for managing mastitis, offering an effective alternative to antibiotics in addressing both bacterial infection and inflammation.
Collapse
Affiliation(s)
- Yipeng Pang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Yongding Ke
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Fructueux Modeste Amona
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Xiaohan Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Zilu Liu
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Junhao Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Yuan Liang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Fang Wang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Yanhong Wang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Xingtang Fang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.
| | - Xi Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.
| | - Chunlei Zhang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.
| |
Collapse
|
35
|
Zhang X, Fang Y, Rong D, Li J, Li Z, Qiu H, Chen Q, Yang J, Wang C, Huang J, Zhao Q, Yang S, Wang H. A novel taxane SB-T-101141 triggers a noncanonical ferroptosis to overcome Paclitaxel resistance of breast cancer via iron homeostasis-related KHSRP. Cell Death Dis 2025; 16:403. [PMID: 40389408 PMCID: PMC12089390 DOI: 10.1038/s41419-025-07710-0] [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: 01/10/2025] [Revised: 04/16/2025] [Accepted: 05/01/2025] [Indexed: 05/21/2025]
Abstract
Acquired multidrug resistance impedes the clinical application of paclitaxel. Here, we disclosed that the taxane SB-T-101141 efficiently contributed to a novel ferroptosis-like cell death of Paclitaxel-resistant and parental breast cancer cells. Functionally, SB-T-101141 facilitated the production of iron and ferrous ions along with reactive oxygen species (ROS), composed of lipid ROS and lipid peroxidation-derived aldehydes, including malonaldehyde (MDA), and glutathione (GSH) depletion. Iron chelators and ROS scavengers significantly attenuated cell death, and the inorganic ROS rendered by SB-T-101141. However, the ferroptosis-associated lipid oxide inhibitors could not block the lipid ROS and cell death triggered by SB-T-101141. Meanwhile, via genome-scale CRISPR-Cas9 screening, we uncovered that SB-T-101141 bound to the KH-type splicing regulatory protein (KHSRP) to inhibit the iron-dependent expression of CDGSH iron sulfur domain 1 (CISD1) associated with iron homeostasis, which consequently led to a novel type of ferroptosis of breast tumors. Moreover, RNA deep sequencing indicated that SB-T-101141 synergistically enhanced the iron-dependent activation of JNK and PERK pathways via KHSRP. Altogether, our results here demonstrate the potential clinical application of SB-T-101141 as a novel ferroptosis inducer in Paclitaxel-resistant breast cancer treatment.
Collapse
Affiliation(s)
- Xiaomei Zhang
- Centre for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Fang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dade Rong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jie Li
- Engineering Research Center of Tibetan Medicine Detection Technology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Zhe Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huidan Qiu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiuxia Chen
- Centre for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Changwei Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Junxiu Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qin Zhao
- Engineering Research Center of Tibetan Medicine Detection Technology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Shulan Yang
- Centre for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Clinical Medical Research Centre for Plateau Gastroenterological Diseases of Xizang Autonomous Region, Xizang Minzu University, Xianyang, China.
| |
Collapse
|
36
|
Zhang S, Liu J, Yang Y, Tao R, Ren X, Zhou X, Liu S. Valproic acid induces ferroptosis and suppresses the proliferation of MDA-MB-231 cells by targeting FDFT1. Front Pharmacol 2025; 16:1540667. [PMID: 40438588 PMCID: PMC12116638 DOI: 10.3389/fphar.2025.1540667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/30/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Valproic acid (VPA) constitutes a branched-chain, short-chain fatty acid that serves as an antiepileptic medication. It has been increasingly recognized that VPA has presented potential anti-tumor properties, including breast cancer. However, the exploration of novel breast cancer treatment methods necessitates a more comprehensive and in-depth understanding of the novel mechanism of VPA inhibition of breast cancer. It has been proven that farnesyl-diphosphate farnesyltransferase 1 (FDFT1) participate in oncogenesis and development of cancers. However, the effect of FDFT1 on breast cancer is still obscure. Thus, it is important to investigate the potential of VPA to trigger ferroptosis in breast cancer cells via targeting FDFT1. Methods In this study, the underlying mechanisms of VPA on ferroptosis in breast cancer cells were explored in vitro and vivo. Initially, the effects of VPA on the proliferation of breast cancer cells were assessed utilizing the Cell Counting Kit-8, cell counting, and colony formation assays. Subsequently, the ferroptosis in breast cancer cells treated with VPA were determined through the use of the Lipid Peroxidation malondialdehyde Assay Kit, reduced glutathione and oxidized glutathione disulfide Assay Kit, flow cytometry, transmission electron microscopy, and western bloting. To explore the impact of VPA in combination with ferrostatin-1, Erastin or RSL3, on MDA-MB-231 cell proliferation and ferroptosis, respective CCK-8, colony formation and WB assays were conducted. Thereafter, we assessed whether VPA facilitated ferroptosis in MDA-MB-231 cells by modulating the expression of FDFT1. Finally, the anti-breast cancer effects of VPA in vivo were validated through a xenograft mouse model, and histological examination via hematoxylin-eosin staining and immunohistochemistry staining were employed to delve into the underlying mechanisms of VPA's inhibitory effects on breast cancer cells in vivo. Results and Discussion The assay outcomes indicated that VPA impedes the proliferation of breast cancer cells. The findings from the ferroptosis index demonstrated that MDA-MB-231 cells are more sensitive to VPA induced ferroptosis than MCF-7 cells. Subsequent to the introduction of ferrostatin-1 (Fer-1), Erastin or RSL3, it was observed that Fer-1 reversed the ferroptosis facilitated by VPA, whereas Erastin or RSL3, in conjunction with VPA, respectively, induced ferroptosis in MDA-MB-231 cells. We revealed that the downregulation of FDFT1 enhanced proliferation and inhibited ferroptosis of MDA-MB-231 cells. Additionally, we discovered that VPA may facilitate ferroptosis in MDA-MB-231 cells by negatively modulating the levels of the solute carrier family 7 member 11 (SLC7A11) protein through the upregulation of FDFT1 expression. In conclusion, this study elucidated that VPA induced the ferroptosis of MDA-MB-231 cells via targeting FDFT1, representing a novel mechanism underlying its efficacy in potentially inhibiting breast cancer.
Collapse
Affiliation(s)
- Shuxian Zhang
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Jiazhuo Liu
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Yisong Yang
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Ran Tao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xin Ren
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xingzhi Zhou
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Shuangping Liu
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| |
Collapse
|
37
|
Wang X, Xiong X. Mitochondrial Reactive Oxygen Species (mROS) Generation and Cancer: Emerging Nanoparticle Therapeutic Approaches. Int J Nanomedicine 2025; 20:6085-6119. [PMID: 40385494 PMCID: PMC12085131 DOI: 10.2147/ijn.s510972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 04/24/2025] [Indexed: 05/20/2025] Open
Abstract
Mitochondrial reactive oxygen species (mROS) are generated as byproducts of mitochondrial oxidative phosphorylation. Changes in mROS levels are involved in tumorigenesis through their effects on cancer genome instability, sustained cancer cell survival, metabolic reprogramming, and tumor metastasis. Recent advances in nanotechnology offer a promising approach for precise regulation of mROS by either enhancing or depleting mROS generation. This review examines the association between dysregulated mROS levels and key cancer hallmarks. We also discuss the potential applications of mROS-targeted nanoparticles that artificially manipulate ROS levels in the mitochondria to achieve precise delivery of antitumor drugs.
Collapse
Affiliation(s)
- Xinyao Wang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
- Queen Mary School of Nanchang University, Nanchang, People’s Republic of China
| | - Xiangyang Xiong
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
38
|
Guo MG, Yang CF, Yuan F, Yang T, Luo PY, He YB, Yang S, Chen F, Li W, Feng ZW. Bioinformatics and experimental validation of ferroptosis-related genes in steroid-induced osteonecrosis of the femoral head. Front Mol Biosci 2025; 12:1578755. [PMID: 40421419 PMCID: PMC12104091 DOI: 10.3389/fmolb.2025.1578755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
Background Steroid-induced osteonecrosis of the femoral head (SONFH) is a progressive condition that causes increasing disability. It is thought to result from reduced blood flow and oxygen levels in the femoral head, with reactive oxygen species (ROS) playing a key role in triggering ferroptosis. However, the role of ferroptosis in SONFH progression remains underexplored. This study aimed to identify and validate key genes associated with ferroptosis in SONFH using bioinformatics. Methods The study analyzed the SONFH dataset GSE123568, which includes data from 30 SONFH patients and 10 controls. Weighted gene co-expression network analysis (WGCNA) was used to identify differentially expressed genes (DEGs) between the SONFH and control groups. Core genes were identified by intersecting DEGs with ferroptosis-related genes retrieved from FerrDb V2. The diagnostic performance of the key genes was assessed using the receiver operating characteristic (ROC) curve, and a predictive nomogram model was developed. Interaction analysis of these genes was conducted to explore their link with immune infiltration. The expression of these genes in bone tissue from SONFH patients was validated. Finally, drug-protein interactions were predicted using the DSigDB database. Results Differential expression analysis identified 384 DEGs, which were significantly involved in inflammatory pathways. WGCNA revealed four key genes after intersecting DEGs with relevant module genes and ferroptosis-related genes. A nomogram model based on these genes demonstrated strong reliability and validity. Immune infiltration analysis showed significant differences between SONFH patients and controls, with notable associations between immune cell infiltration and the expression of the four core genes. Validation through quantitative real-time PCR (qRT-PCR) and Western blot confirmed that the expression of GCLC, GABARAPL2, CISD2, and NCOA4 was significantly lower in SONFH bone tissue compared to controls (P < 0.05). Additionally, potential therapeutic drugs targeting these genes, including Diethyl sulfate, Meloxicam, and NIMUSTINE, were predicted. Conclusion This study identifies GABARAPL2, CISD2, NCOA4, and GCLC as potential diagnostic biomarkers associated with immune cell infiltration in SONFH, offering new insights for future research and clinical applications.
Collapse
Affiliation(s)
- Ming-gang Guo
- Department of Orthopaedics, Beijing Anzhen Nanchong Hospital, Capital Medical University and Nanchong Central Hospital, Nanchong, China
| | - Chen-fei Yang
- School of Nursing, North Sichuan Medical College, Nanchong, China
| | - Fa Yuan
- Department of Orthopaedics, Beijing Anzhen Nanchong Hospital, Capital Medical University and Nanchong Central Hospital, Nanchong, China
| | - Tao Yang
- Department of Orthopaedics, Nanjiang County People’s Hospital, Bazhong, China
| | - Ping-yuan Luo
- Department of Orthopaedics, Nanjiang County People’s Hospital, Bazhong, China
| | - Yu-bai He
- Department of Orthopaedics, Nanjiang County People’s Hospital, Bazhong, China
| | - Shuan Yang
- Department of Orthopaedics, Nanjiang County People’s Hospital, Bazhong, China
| | - Feng Chen
- Department of Orthopaedics, Beijing Anzhen Nanchong Hospital, Capital Medical University and Nanchong Central Hospital, Nanchong, China
| | - Wei Li
- Department of Orthopaedics, Beijing Anzhen Nanchong Hospital, Capital Medical University and Nanchong Central Hospital, Nanchong, China
| | - Zhi-wei Feng
- Department of Orthopaedics, Beijing Anzhen Nanchong Hospital, Capital Medical University and Nanchong Central Hospital, Nanchong, China
| |
Collapse
|
39
|
Wang T, Liu X, Feng X, Zhang Z, Lv R, Feng W, Zhou Y, Liao X, Tang H, Xu M. GPX4 degradation contributes to heat stress-induced liver injury via chaperone-mediated autophagy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119988. [PMID: 40368268 DOI: 10.1016/j.bbamcr.2025.119988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/19/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Heat stress (HS) is a significant health concern that adversely affects both human and animal health, particularly impacting liver function due to its central metabolic role. This study investigated the mechanisms underlying HS-induced liver injury, focusing on the role of ferroptosis, an iron-dependent form of cell death characterized by lipid peroxidation and cellular iron accumulation. Using mouse and cellular HS models, the results demonstrated that HS induced liver injury through ferroptosis, as evidenced by increased levels of malondialdehyde (MDA), oxidized glutathione (GSSG), and iron, alongside decreased glutathione (GSH) and glutathione peroxidase 4 (GPX4) expression. The ferroptosis inhibitor Ferrostatin-1 (Fer-1) effectively mitigated HS-induced liver damage, reducing oxidative stress and restoring GPX4 levels. Furthermore, HS promoted the lysosomal degradation of GPX4 via the chaperone-mediated autophagy (CMA) pathway, which was regulated by heat shock cognate protein 70 (HSC70) and lysosome-associated membrane protein 2A (LAMP2A). Knockdown of LAMP2A in hepatocytes significantly suppressed HS-induced GPX4 degradation, confirming the critical role of CMA in this process. Inhibition of CMA using Apoptozole, an HSC70 inhibitor, or Bafilomycin A1 (Baf-A1), a lysosomal inhibitor, further attenuated HS-induced ferroptosis and liver injury. These findings highlight the critical role of CMA-mediated GPX4 degradation in HS-induced ferroptosis and liver injury, providing potential therapeutic targets for mitigating HS-related liver damage.
Collapse
Affiliation(s)
- Ting Wang
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xiao Liu
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xinyu Feng
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Zhenyu Zhang
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ruiyi Lv
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Wenhong Feng
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yukun Zhou
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xueyu Liao
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Haoming Tang
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ming Xu
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.
| |
Collapse
|
40
|
Xie L, Zhang Y, Niu X, Kang Y, Li W, Yao J. A comprehensive bibliometric analysis of ferroptosis in tumor resistance: development and emerging trends. Front Immunol 2025; 16:1580222. [PMID: 40416987 PMCID: PMC12098629 DOI: 10.3389/fimmu.2025.1580222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/21/2025] [Indexed: 05/27/2025] Open
Abstract
Background Ferroptosis is a regulated form of cell death characterized by iron dependency, lipid peroxidation, and oxidative stress. Since its discovery in 2012, ferroptosis has attracted significant interest for its potential to counteract tumor resistance across various therapeutic modalities, including chemotherapy, radiotherapy, immunotherapy, and targeted therapy. Despite notable progress, a systematic understanding of its underlying molecular mechanisms and translational potential remains underdeveloped, thus necessitating a comprehensive bibliometric analysis. Methods We employed bibliometric tools, including VOSviewer, CiteSpace, and bibliometric.com, to analyze 2,663 articles related to ferroptosis and tumor resistance indexed in the Web of Science Core Collection from 2014 to 2024. The analysis included co-occurrence, co-citation, and clustering techniques to explore trends, influential keywords, prominent journals, leading institutions, and key contributors. Citation burst detection and temporal analysis were used to uncover emerging research hotspots and track the field's evolution. Results Over the past decade, the volume of publications in this field has grown rapidly, with China and the United States leading in both research output and academic influence. Notable institutions such as Central South University and Fudan University contributed significantly, while Kang Rui and Tang Daolin emerged as prolific authors. Key research hotspots identified include oxidative stress, tumor microenvironment, and nanomedicine, with emerging themes such as immunotherapy and autophagy gaining prominence. Temporal trends indicated a shift from mechanistic studies toward translational applications, emphasizing the integration of ferroptosis in clinical strategies to address tumor resistance. Conclusions This bibliometric analysis highlights ferroptosis as a rapidly evolving field with significant contributions to understanding tumor resistance mechanisms. The identification of emerging themes and promising research directions offers valuable insights for future investigations and clinical applications of ferroptosis in overcoming tumor resistance.
Collapse
Affiliation(s)
| | | | | | | | | | - Jun Yao
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
41
|
Zhao J, Yin Y, Liu M, Lu Y, Cao J, Qi X, Wu L, Shen S. Ferritin/Ferroportin-Regulating Nanoparticles Boosting Intracellular Free Iron for Enhanced Ferrotherapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26445-26454. [PMID: 40277408 DOI: 10.1021/acsami.5c04135] [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: 04/26/2025]
Abstract
Ferroptosis therapy efficacy of cancers suffers from relatively low concentrations of intracellular free iron ions due to the efficient regulation of iron through storage in ferritin and efflux via ferroportin (FPN). In this study, a ferritin/ferroportin-hijacking nanoplatform (Fe3O4-ART@MM-Hep) containing artemisinin (ART) and hepcidin (Hep) is fabricated to boost intracellular free iron ions and induce reactive oxygen species (ROS) storm. Once the tumor site is reached, the hepcidin targeted binds to FPN and triggers the internalization and degradation of FPN, blocking the efflux of intracellular iron ions. Meanwhile, artemisinin induces lysosomal degradation of ferritin, liberating the endogenous iron. Combined with exogenous iron supplemented by Fe3O4, the nanoplatform facilities the generation of ROS. What's more, the released Fe2+ catalyzes artemisinin to generate carbon-centered free radicals, further enhancing tumor killing ability. All of the above strategies trigger an ROS storm in tumor cells and indicate a promising platform for high-performance ferrotherapy.
Collapse
Affiliation(s)
- Junsheng Zhao
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yanzhao Yin
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Mengxiao Liu
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Ying Lu
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jin Cao
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xueyong Qi
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Lin Wu
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Song Shen
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| |
Collapse
|
42
|
Xu M, Xu B. Protein lipidation in the tumor microenvironment: enzymology, signaling pathways, and therapeutics. Mol Cancer 2025; 24:138. [PMID: 40335986 PMCID: PMC12057185 DOI: 10.1186/s12943-025-02309-7] [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: 11/27/2024] [Accepted: 03/18/2025] [Indexed: 05/09/2025] Open
Abstract
Protein lipidation is a pivotal post-translational modification that increases protein hydrophobicity and influences their function, localization, and interaction network. Emerging evidence has shown significant roles of lipidation in the tumor microenvironment (TME). However, a comprehensive review of this topic is lacking. In this review, we present an integrated and in-depth literature review of protein lipidation in the context of the TME. Specifically, we focus on three major lipidation modifications: S-prenylation, S-palmitoylation, and N-myristoylation. We emphasize how these modifications affect oncogenic signaling pathways and the complex interplay between tumor cells and the surrounding stromal and immune cells. Furthermore, we explore the therapeutic potential of targeting lipidation mechanisms in cancer treatment and discuss prospects for developing novel anticancer strategies that disrupt lipidation-dependent signaling pathways. By bridging protein lipidation with the dynamics of the TME, our review provides novel insights into the complex relationship between them that drives tumor initiation and progression.
Collapse
Affiliation(s)
- Mengke Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China.
| |
Collapse
|
43
|
Xiong Z, Liao Y, Zhang Z, Wan Z, Liang S, Guo J. Molecular Insights into Oxidative-Stress-Mediated Cardiomyopathy and Potential Therapeutic Strategies. Biomolecules 2025; 15:670. [PMID: 40427563 PMCID: PMC12108637 DOI: 10.3390/biom15050670] [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: 03/27/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiomyopathies comprise a heterogeneous group of cardiac disorders characterized by structural and functional abnormalities in the absence of significant coronary artery disease, hypertension, valvular disease, or congenital defects. Major subtypes include hypertrophic, dilated, arrhythmogenic, and stress-induced cardiomyopathies. Oxidative stress (OS), resulting from an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, has emerged as a key contributor to the pathogenesis of these conditions. ROS-mediated injury drives inflammation, protease activation, mitochondrial dysfunction, and cardiomyocyte damage, thereby promoting cardiac remodeling and functional decline. Although numerous studies implicate OS in cardiomyopathy progression, the precise molecular mechanisms remain incompletely defined. This review provides an updated synthesis of current findings on OS-related signaling pathways across cardiomyopathy subtypes, emphasizing emerging therapeutic targets within redox-regulatory networks. A deeper understanding of these mechanisms may guide the development of targeted antioxidant strategies to improve clinical outcomes in affected patients.
Collapse
Affiliation(s)
- Zhenyu Xiong
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Yuanpeng Liao
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhaoshan Zhang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhengdong Wan
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
| | - Sijia Liang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiawei Guo
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
44
|
Jing L, Xiao W, Hu Z, Liu X, Yuan M. A Systematic Review of Nanoparticle-Mediated Ferroptosis in Glioma Therapy. Int J Nanomedicine 2025; 20:5779-5797. [PMID: 40351706 PMCID: PMC12065465 DOI: 10.2147/ijn.s523008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Glioma, a highly malignant central nervous system tumor, exhibits aggressive invasiveness, extensive infiltration, and poor prognosis. Conventional treatments such as surgery, radiotherapy, and chemotherapy are hindered by limitations including the inability to overcome the blood-brain barrier (BBB), drug resistance, and high recurrence rates. Ferroptosis induced by nanoparticle-based systems offers an innovative strategy for glioma therapy by efficiently traversing the BBB, precisely delivering ferroptosis inducers, enhancing tumor accumulation, and enabling stimuli-responsive drug release. These features collectively improve the induction efficiency of ferroptosis in glioma cells. Various nanoplatforms, including inorganic nanoparticles, biomimetic carriers, and polymer-based systems, have demonstrated potential in crossing the BBB, inducing ferroptosis, and suppressing glioma progression. These systems enhance reactive oxygen species generation, deplete glutathione, and disrupt tumor microenvironment defense mechanisms, achieving synergistic therapeutic effects. The integration of ferroptosis with nanotechnology is emerging as a promising, non-invasive strategy for the treatment of gliomas, offering substantial therapeutic potential.
Collapse
Affiliation(s)
- Lin Jing
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Wenguang Xiao
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Zhouxing Hu
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Xu Liu
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Mingqing Yuan
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| |
Collapse
|
45
|
Chen C, Wang L, Cui XF, Shang XY, Bai SH, Li L, Wang N, Han ZG. SCARA5 deficiency inhibits ferroptosis via regulating iron homeostasis and results in sorafenib resistance in hepatocellular carcinoma. Cell Signal 2025; 129:111656. [PMID: 39954713 DOI: 10.1016/j.cellsig.2025.111656] [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: 10/30/2024] [Revised: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
SCARA5 (Scavenger Receptor Class A Member 5), a member of scavenger receptor class A, is a type II transmembrane protein. Previous studies, including our own, have suggested that SCARA5 acts as a tumor suppressor in various cancers. Additionally, SCARA5 has been identified as a ferritin receptor that facilitates iron delivery independent of transferrin. However, it remains unclear whether ferroptosis is involved in the tumor-suppressive function of SCARA5 in hepatocellular carcinoma (HCC). In this study, we found that SCARA5-deficient cells, including mouse embryonic fibroblasts (MEFs) and HCC cells, exhibited reduced sensitivity to ferroptosis induced by erastin and RSL3. We measured the cell viability, cellular reactive oxygen species (ROS), lipid ROS, malondialdehyde (MDA) and ferrous iron concentration to assess the role of SCARA5 in ferroptosis. Mechanistically, we confirmed that SCARA5 might enhance the intracellular availability of bioactive ferrous iron by promoting autophagic degradation of the major iron storage protein ferritin. Furthermore, we found that SCARA5 deficiency contributed to the resistance of HCC cells to sorafenib, a therapeutic agent for HCC, possibly by inhibiting ferroptosis. Collectively, our study revealed the role of SCARA5 in regulating ferroptosis, providing a profound understanding of sorafenib resistance in HCC systemic therapy.
Collapse
Affiliation(s)
- Cong Chen
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lan Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiao-Fang Cui
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xu-Yang Shang
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shi-Hao Bai
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lin Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Na Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ze-Guang Han
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
46
|
Jian PA, Yang TN, Wang YX, Ma XY, Huang NN, Ren YF, Yuan SH, Li JL, Wang CC, Li XN. Lycopene, a natural plant extract, alleviates atrazine-induced ferroptosis in hepatocytes by activating cytochrome P450 oxidoreductase. Int J Biol Macromol 2025; 308:142311. [PMID: 40139611 DOI: 10.1016/j.ijbiomac.2025.142311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Atrazine (ATZ) and diaminochlorotriazine (DACT) accumulation poses liver health risks in animals and humans. Lycopene (LYC), a carotenoid found in red plants and fruits, exhibits potent antioxidant effects. This study explores the interaction between LYC and ATZ in mouse hepatocyte ferroptosis and the potential regulatory role of Cytochrome P450 oxidoreductase (CYPOR) in this process. Male mice were exposed to ATZ (50 mg/kg or 200 mg/kg) and/or LYC (5 mg/kg) by gavage for 21 days. In vitro experiments, a mouse hepatocyte cell line (AML12) was exposed to DACT (200 μM) and/or LYC (2 μM) for 12 h with or without small interfering RNA treatment. We found that both ATZ and DACT promoted CYPOR expression and caused liver injury. ATZ/DACT promotes Fe2+ accumulation and lipid peroxidation, ultimately leading to Ferroptosis in mouse hepatocytes. However, LYC alleviated ATZ/DACT-induced Ferroptosis by inhibiting CYPOR. The CYPOR knockdown resulted in the blockage of ATZ/DACT-induced ferroptosis, while the alleviation of ferroptosis by LYC was further enhanced. Thus, CYPOR can regulate ferroptosis in mouse hepatocytes and is a novel target for the treatment of hepatocyte ferroptosis-related diseases. Lycopene can be used as a functional dietary supplement to scavenge ferroptosis and reduce chronic liver disease.
Collapse
Affiliation(s)
- Ping-An Jian
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Tian-Ning Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yu-Xiang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiang-Yu Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Ning-Ning Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yi-Fei Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shi-Hao Yuan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China
| | - Chi-Chiu Wang
- Department of Obstetrics & Gynaecology, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, The Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong.
| | - Xue-Nan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Department of Obstetrics & Gynaecology, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, The Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
47
|
Bojuan L, Youdong Z, Lei W, Lixin X, Jinyang M. Oleanolic Acid Alleviates Neuronal Ferroptosis in Subarachnoid Hemorrhage by Inhibiting KEAP1-Nrf2 and NF-κB Pathways. Drug Dev Res 2025; 86:e70105. [PMID: 40358968 DOI: 10.1002/ddr.70105] [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: 09/09/2024] [Revised: 03/10/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025]
Abstract
Oleanolic acid (OA) is a pentacyclic triterpenoid compound, and we previously report that it ameliorates neurological injury in subarachnoid hemorrhage (SAH) model. However, the underlying mechanism is not clear. The aim of this study was to explore the effect and mechanism of OA on SAH. In this study, network pharmacology was applied to screen the targets of OA in SAH treatment. Based on these targets, protein-protein interaction network was constructed, and k-means cluster analysis was used to screen the core targets of OA in SAH treatment. In vitro SAH model was constructed with hemin-induced neuron HT22 and microglia BV2. Then cell counting Kit 8, flow cytometry, western blot, qPCR were performed to evaluate the effects of OA on neurons and microglia. 93 targets were identified as the core targets of OA in SAH treatment. Notably, these targets are closely related to neuroinflammation and oxidative stress responses. OA had good binding activity with KEAP1, NFKB1 and IKBA. OA significantly alleviated the inhibitory effect of hemin on HT22 cell viability. OA significantly inhibited the expression of CD86, promoted the expression of CD206, and promoted the transformation of microglia from M1 type to M2 type. Additionally, OA could inhibit the activation of NF-κB and KEAP1/Nrf2 pathways. In conclusion, OA ameliorates inflammatory response, oxidative stress and ferroptosis in SAH, and suppresses neuronal injury by inhibiting NF-κB and KEAP1/Nrf2 pathways.
Collapse
Affiliation(s)
- Lang Bojuan
- Department of Pathology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, PR China
| | - Zhou Youdong
- Department of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, PR China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Wang Lei
- Department of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, PR China
| | - Xue Lixin
- Department of Neurosurgery, Zhijiang Branch of Yichang Central People's Hospital, Yichang, PR China
| | - Ma Jinyang
- Department of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, PR China
| |
Collapse
|
48
|
Khodadadi H, Łuczyńska K, Winiarczyk D, Leszczyński P, Taniguchi H. NFE2L1 as a central regulator of proteostasis in neurodegenerative diseases: interplay with autophagy, ferroptosis, and the proteasome. Front Mol Neurosci 2025; 18:1551571. [PMID: 40375958 PMCID: PMC12078313 DOI: 10.3389/fnmol.2025.1551571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/14/2025] [Indexed: 05/18/2025] Open
Abstract
Maintaining proteostasis is critical for neuronal health, with its disruption underpinning the progression of neurodegenerative diseases such as Alzheimer's, Parkinson's, and Huntington's diseases. Nuclear Factor Erythroid 2-Related Factor 1 (NFE2L1) has emerged as a key regulator of proteostasis, integrating proteasome function, autophagy, and ferroptosis to counteract oxidative stress and protein misfolding. This review synthesizes current knowledge on the role of NFE2L1 in maintaining neuronal homeostasis, focusing on its mechanisms for mitigating proteotoxic stress and supporting cellular health, offering protection against neurodegeneration. Furthermore, we discuss the pathological implications of NFE2L1 dysfunction and explore its potential as a therapeutic target. By highlighting gaps in the current understanding and presenting future research directions, this review aims to elucidate NFE2L1's role in advancing treatment strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Hossein Khodadadi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Kamila Łuczyńska
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
- The Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, Warsaw, Poland
| | - Dawid Winiarczyk
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Paweł Leszczyński
- Department of Stem Cell Bioengineering Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Hiroaki Taniguchi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
- African Genome Center, University Mohammed VI Polytechnic (UM6P), Ben Guerir, Morocco
| |
Collapse
|
49
|
Saha A, Islam MM, Kumar R, Ismail AM, Garcia E, Gullapali RR, Chodosh J, Rajaiya J. Virus and cell specific HMGB1 secretion and subepithelial infiltrate formation in adenovirus keratitis. PLoS Pathog 2025; 21:e1013184. [PMID: 40367285 PMCID: PMC12101768 DOI: 10.1371/journal.ppat.1013184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 05/23/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
A highly contagious infection caused by human adenovirus species D (HAdV-D), epidemic keratoconjunctivitis (EKC) results in corneal subepithelial infiltration (SEI) by leukocytes, the hallmark of the infection. To date, the pathogenesis of corneal SEI formation in EKC is unresolved. HMGB1 (high-mobility group box 1 protein) is an alarmin expressed in response to infection and a marker of sepsis. Earlier studies using a different adenovirus species, HAdV-C, showed retention of HMGB1 in the infected cell nucleus by adenovirus protein VII, enabling immune evasion. Here, using HAdV-D we show cell-specific HMGB1 secretion by infected cells, and provide an HAdV-D specific mechanism for SEI formation in EKC. HMGB1 was secreted only upon infection of human corneal epithelial cells, not from other cell types, and only upon infection by HAdV-D types associated with EKC. Acetylated HMGB1 translocation from the nucleus to the cytoplasm, then to the extracellular milieu, was tightly controlled by CRM1 and LAMP1, respectively. Primary stromal cells when stimulated by rHMGB1 expressed proinflammatory chemokines. In a novel 3D culture system in tune with the architecture of the cornea, HMGB1 released by infected corneal epithelial cells induced leukocytic infiltrates either directly and/or indirectly via stimulated stromal cells, which together explains SEI formation in EKC.
Collapse
Affiliation(s)
- Amrita Saha
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Mohammad Mirazul Islam
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Ophthalmology and Visual Sciences, University of Ophthalmology and Visual Sciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Rahul Kumar
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Ashrafali Mohamed Ismail
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Emanuel Garcia
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Rama R. Gullapali
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
- Department of Ophthalmology and Visual Sciences, University of Ophthalmology and Visual Sciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Jaya Rajaiya
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| |
Collapse
|
50
|
Zhao Y, Wang XQ, Liu RQ, Jiang FW, Wang JX, Chen MS, Zhang H, Cui JG, Chang YH, Li JL. SLC7A11 as a therapeutic target to attenuate phthalates-driven testosterone level decline in mice. J Adv Res 2025; 71:369-381. [PMID: 38797476 DOI: 10.1016/j.jare.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024] Open
Abstract
INTRODUCTION Phthalates exposure is a major public health concern due to the accumulation in the environment and associated with levels of testosterone reduction, leading to adverse pregnancy outcomes. However, the relationship between phthalate-induced testosterone level decline and ferroptosis remains poorly defined. OBJECTIVES Herein, we aimed to explore the mechanisms of phthalates-induced testosterone synthesis disorder and its relationship to ferroptosis. METHODS We conducted validated experiments in vivo male mice model and in vitro mouse Leydig TM3 cell line, followed by RNA sequencing and metabolomic analysis. We evaluated the levels of testosterone synthesis-associated enzymes and ferroptosis-related indicators by using qRT-PCR and Western blotting. Then, we analyzed the lipid peroxidation, ROS, Fe2+ levels and glutathione system to confirm the occurrence of ferroptosis. RESULTS In the present study, we used di (2-ethylhexyl) phthalate (DEHP) to identify ferroptosis as the critical contributor to phthalate-induced testosterone level decline. It was demonstrated that DEHP caused glutathione metabolism and steroid synthesis disorders in Leydig cells. As the primary metabolite of DEHP, mono-2-ethylhexyl phthalate (MEHP) triggered testosterone synthesis disorder accompanied by a decrease in the expression of solute carri1er family 7 member 11 (SLC7A11) protein. Furthermore, MEHP synergistically induced ferroptosis with Erastin through the increase of intracellular and mitochondrial ROS, and lipid peroxidation production. Mechanistically, overexpression of SLC7A11 counteracts the synergistic effect of co-exposure to MEHP-Erastin. CONCLUSION Our research results suggest that MEHP does not induce ferroptosis but synergizes Erastin-induced ferroptosis. These findings provide evidence for the role of ferroptosis in phthalates-induced testosterone synthesis disorder and point to SLC7A11 as a potential target for male reproductive diseases. This study established a correlation between ferroptosis and phthalates cytotoxicity, providing a novel view point for mitigating the issue of male reproductive disease and "The Global Plastic Toxicity Debt".
Collapse
Affiliation(s)
- Yi Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xue-Qi Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Rui-Qi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Fu-Wei Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jia-Xin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Ming-Shan Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jia-Gen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yuan-Hang Chang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|