1
|
Gong L, Wu L, Zhao S, Xiao S, Chu X, Zhang Y, Li F, Li S, Yang H, Jiang P. Epigenetic regulation of ferroptosis in gastrointestinal cancers (Review). Int J Mol Med 2025; 55:93. [PMID: 40242977 PMCID: PMC12045471 DOI: 10.3892/ijmm.2025.5534] [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: 11/13/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Ferroptosis is a type of iron‑dependent cell death characterized by excessive lipid peroxidation and may serve as a potential therapeutic target in cancer treatment. While the mechanisms governing ferroptosis continue to be explored and elucidated, an increasing body of research highlights the significant impact of epigenetic modifications on the sensitivity of cancer cells to ferroptosis. Epigenetic processes, such as DNA methylation, histone modifications and non‑coding RNAs, have been identified as key regulators that modulate the expression of ferroptosis‑related genes. These alterations can either enhance or inhibit the sensitivity of gastrointestinal cancer (GIC) cells to ferroptosis, thereby affecting the fate of GICs. Drugs that target epigenetic markers for advanced‑stage cancer have shown promising results in enhancing ferroptosis and inhibiting tumor growth. This review explores the intricate relationship between epigenetic regulation and ferroptosis in GICs. Additionally, the potential of leveraging epigenetic modifications to trigger ferroptosis in GICs is investigated. This review highlights the importance of further research to elucidate the specific mechanisms underlying epigenetic control of ferroptosis and to advance the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Linqiang Gong
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Linlin Wu
- Oncology Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shiyuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| | - Shuai Xiao
- Department of Intensive Care Medicine, Tengzhou Central People's Hospital, Jining Medical University, Tengzhou, Shandong 277500, P.R. China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
| | - Yazhou Zhang
- Department of Foot and Ankle Surgery, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Fengfeng Li
- Neurosurgery Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shuhui Li
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Hui Yang
- Department of Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| |
Collapse
|
2
|
Dai X, Zheng Y, Cui J, Zeng Y, Yang B, Zhang Z. Nanodrug delivery systems targeting ferroptosis as an innovative therapeutic approach for Rheumatoid Arthritis. Mater Today Bio 2025; 32:101804. [PMID: 40343168 PMCID: PMC12059336 DOI: 10.1016/j.mtbio.2025.101804] [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: 02/13/2025] [Revised: 04/02/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
Rheumatoid Arthritis (RA) is a chronic inflammatory disease characterized by joint inflammation, progressive cartilage degradation, and bone erosion. Recent research has implicated ferroptosis not only in autoimmune hepatitis but also in the pathogenesis and progression of autoimmune disorders like RA. Consequently, numerous therapeutic strategies have begun to target the ferroptosis pathway, particularly in the design and development of nanodrug delivery systems (NDDSs). While previous reviews have comprehensively discussed the mechanisms of ferroptosis, related signaling pathways, and NDDS materials, recent studies have further elucidated the interplay between ferroptosis and various metabolic pathways, providing a robust theoretical basis for the design of NDDS-based ferroptosis strategies. This review focuses on investigating the role of ferroptosis in the development of RA, aiming to elucidate how targeting ferroptosis can offer novel therapeutic concepts and potential treatments for RA patients. Specifically, it summarizes the design strategies of ferroptosis-based NDDSs via different pathways and highlights the feasibility of RA treatment regimens based on the ferroptosis mechanism. Furthermore, the review critically discusses the current limitations of NDDSs and offers perspectives on future research directions in this field.
Collapse
Affiliation(s)
- Xiaolin Dai
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Yu Zheng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, 646000, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, PR China
- Sichuan-Chongqing Joint Key Laboratory of Metabolic Vascular Diseases, Luzhou, 646000, PR China
| | - Jianrong Cui
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Yuqi Zeng
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Bo Yang
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Zhanlin Zhang
- Irradiation Preservation and Effect Key Laboratory of Sichuan Province, School of Bioscience and Technology, Chengdu Medical College, Chengdu, 610500, PR China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, PR China
| |
Collapse
|
3
|
Zhang Z, Yang J, Zhou Q, Zhong S, Liu J, Zhang X, Chang X, Wang H. The cGAS-STING-mediated ROS and ferroptosis are involved in manganese neurotoxicity. J Environ Sci (China) 2025; 152:71-86. [PMID: 39617588 DOI: 10.1016/j.jes.2024.05.003] [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: 01/31/2024] [Revised: 04/12/2024] [Accepted: 05/01/2024] [Indexed: 12/12/2024]
Abstract
Manganese (Mn) has been characterized as an environmental pollutant. Excessive releases of Mn due to human activities have increased Mn levels in the environment over the years, posing a threat to human health and the environment. Long-term exposure to high concentrations of Mn can induce neurotoxicity. Therefore, toxicological studies on Mn are of paramount importance. Mn induces oxidative stress through affecting the level of reactive oxygen species (ROS), and the overabundance of ROS further triggers ferroptosis. Additionally, Mn2+ was found to be a novel activator of the cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway in the innate immune system. Thus, we speculate that Mn exposure may promote ROS production by activating the cGAS-STING pathway, which further induces oxidative stress and ferroptosis, and ultimately triggers Mn neurotoxicity. This review discusses the mechanism between Mn-induced oxidative stress and ferroptosis via activation of the cGAS-STING pathway, which may offer a prospective direction for future in-depth studies on the mechanism of Mn neurotoxicity.
Collapse
Affiliation(s)
- Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
4
|
Jia SZ, Li Y, Xu XW, Huang YP, Deng XY, Zhang ZH, Song GL. Selenoprotein K Confers Protection against Iron Dyshomeostasis-Related Neurotoxicity by Regulating the Palmitoylation of TfR-1. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12233-12246. [PMID: 40296316 DOI: 10.1021/acs.jafc.4c08266] [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: 04/30/2025]
Abstract
Selenoprotein K (SELENOK), a protein residing in the endoplasmic reticulum (ER), is modulated by dietary selenium and is expressed at elevated levels in neurons. SELENOK has been shown to participate in cellular antioxidant activity and posttranslational palmitoylation. This study presents both in vivo and in vitro evidence that SELENOK deficiency reduces the palmitoylation of TfR-1, thereby impairing transferrin transport and ultimately leading to a decrease in the intracellular iron content, impaired mitochondrial respiratory chain activity and decreased ATP production. Remarkably, restoring SELENOK levels significantly enhanced TfR-1 palmitoylation, increased intracellular iron levels, and restored mitochondrial function, thus ameliorating cognitive deficits in 7 month-old SELENOK knockout mice. Consistent with these findings, iron supplementation also improved mitochondrial function by elevating intracellular iron levels, thereby improving cognitive deficits in 7 month-old SELENOK knockout mice. Therefore, SELENOK exerts its neuroprotective effect by regulating the palmitoylation of TfR-1 to maintain iron homeostasis, thereby protecting mitochondrial function in neurons.
Collapse
Affiliation(s)
- Shi-Zheng Jia
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Yu Li
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Xin-Wen Xu
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Yan-Ping Huang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Xiao-Yi Deng
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Zhong-Hao Zhang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518060, China
| | - Guo-Li Song
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518060, China
| |
Collapse
|
5
|
Zeng B, Zhang C, Liang Y, Huang J, Li D, Liu Z, Liao H, Yang T, Liu M, Zou C, Liu D, Qin B. Single-cell RNA sequencing highlights a significant retinal Müller glial population in dry age-related macular degeneration. iScience 2025; 28:112464. [PMID: 40343286 PMCID: PMC12059717 DOI: 10.1016/j.isci.2025.112464] [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: 08/05/2024] [Revised: 12/21/2024] [Accepted: 04/14/2025] [Indexed: 05/11/2025] Open
Abstract
The main challenge in dissecting the cells and pathways involved in the pathogenesis of age-related macular degeneration (AMD) is the highly heterogeneous and dynamic nature of the retinal microenvironment. This study aimed to describe the comprehensive landscape of the dry AMD (dAMD) model and identify the key cell cluster contributing to dAMD. We identified a subset of Müller cells that express high levels of Sox2, which play crucial roles in homeostasis and neuroprotection in both mouse models of AMD and patients with dAMD. Additionally, the number of Sox2+ Müller cells decreased significantly during the progression of AMD, indicating these cells were damaged and underwent cell death. Interestingly, ferroptosis and apoptosis were identified as contributors to the damage of Sox2+ Müller cells. Our findings are potentially valuable not only for advancing the current understanding of dAMD progression but also for the development of treatment strategies through the protection of Müller cells.
Collapse
Affiliation(s)
- Bing Zeng
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Chuanhe Zhang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Yifan Liang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Jianguo Huang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Deshuang Li
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Ziling Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Hongxia Liao
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Tedu Yang
- Shenzhen Shendong Aier Eye Hospital, Shenzhen, China
| | - Muyun Liu
- National Engineering Research Center of Foundational Technologies for CGT Industry, Shenzhen Kenuo Medical Laboratory, Shenzhen, Guangdong, China
| | - Chang Zou
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, P.R. China
| | - Dongcheng Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
- Aier School of Ophthalmology, Central South University, Changsha, China
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
- Aier School of Ophthalmology, Central South University, Changsha, China
| |
Collapse
|
6
|
Ding T, Song M, Wu Y, Li Z, Zhang S, Fan X. Schisandrin B ameliorates Alzheimer's disease by suppressing neuronal ferroptosis and ensuing microglia M1 polarization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156780. [PMID: 40382817 DOI: 10.1016/j.phymed.2025.156780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/24/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder characterized by neuronal damage, with poor prognosis and limited therapeutic options. Inhibition of neuronal ferroptosis has shown promise as a potential treatment for AD. Schisandrin B (Sch B), a major active component of Schisandra chinensis, exhibits potential neuroprotective effects. However, whether Sch B inhibits neuronal ferroptosis remains unclear. PURPOSE To investigate the mechanisms underlying the effects of Sch B on the GSK3β/Nrf2/GPX4 and FSP1 signaling pathways, which are the suppression of neuronal ferroptosis and the potential therapeutic intervention in AD. METHODS We employed the 3 × Tg mouse model in vivo, and utilized the erastin-induced ferroptosis model in SH-SY5Y/APP695swe cells in vitro. Nissl staining was conducted to facilitate histopathological assessment. Assessment of neuronal ferroptosis was performed utilizing a lipid peroxidation and ferroptosis marker assay kit. Furthermore, bioinformatic analysis was executed with the application of the GEO database. Immunofluorescence and Western blot analyses were performed to quantify protein expression levels within the cellular context. ELISA was utilized to determine cytokine concentrations within the supernatant of cell cultures. RT-PCR was executed to evaluate mRNA expression levels. RESULTS Sch B suppresses the activation of GSK3β, modulating the Nrf2/GPX4 signaling pathway and consequently inhibiting ferroptosis in neurons, which results in amelioration of cognitive impairment and pathological damage in 3 × Tg mice. Sch B also inhibits GSK3β activation, thereby modulating the Nrf2/GPX4 signaling pathway to prevent erastin-induced ferroptosis in SH-SY5Y695swe cells in vitro. Furthermore, Sch B modulates FSP1, enhancing its synergistic interaction with the GSK3β/Nrf2/GPX4 pathway to suppress neuronal ferroptosis. Sch B can also inhibit TNF-α release from neurons undergoing ferroptosis, thus impeding the activation of M1-type microglia, suggesting a multifaceted neuroprotective strategy against neuroinflammatory processes. CONCLUSION Sch B modulates the GSK3β/Nrf2/GPX4 pathway in conjunction with FSP1 to inhibit neuronal ferroptosis and the subsequent microglial M1 polarization mediated by neuronal ferroptosis, thereby improving cognitive impairment and pathological damage in AD.
Collapse
Affiliation(s)
- Tao Ding
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Meiying Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yongshi Wu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhu Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
7
|
Guo S, Zhang Q, Ge H, Wang H. Baicalin plays a protective role by regulating ferroptosis in multiple diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4837-4849. [PMID: 39661143 DOI: 10.1007/s00210-024-03704-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
Ferroptosis is a new kind of cell death discovered in recent years, usually accompanied by a large number of lipid peroxidation and iron accumulation in the process of cell death. Ferroptosis has been proven to play an important role in various diseases, including ischemic reperfusion injury, cancer, and neurodegeneration. Therefore, the regulation of ferroptosis will have a vital impact on the occurrence and development of diseases. Baicalin is a flavonoid compound extracted and isolated from the dried roots of Scutellaria baicalensis Georgi, a plant in the family Lamiaceae. It has various biological activities such as antioxidant, anti-proliferative, anti-inflammatory, anti-thrombotic, and regulates apoptosis and ferroptosis. Recently, increasing evidence indicates that baicalin regulation of ferroptosis is involved in multiple diseases. However, the relevant mechanisms are not yet fully understood. Here, we summarized the role of baicalin regulation of ferroptosis in different kinds of diseases, and conducted an in-depth analysis of the relevant mechanisms, hoping to provide the theoretical references for future related researches.
Collapse
Affiliation(s)
- Shiyun Guo
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Qi Zhang
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Hangwei Ge
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
8
|
Zhao N, Li S, Wu H, Wei D, Pu N, Wang K, Liu Y, Tao Y, Song Z. Ferroptosis: An Energetic Villain of Age-Related Macular Degeneration. Biomedicines 2025; 13:986. [PMID: 40299661 PMCID: PMC12024642 DOI: 10.3390/biomedicines13040986] [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: 02/16/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Iron homeostasis plays an important role in maintaining cellular homeostasis; however, excessive iron can promote the production of reactive oxygen species (ROS). Ferroptosis is iron-dependent programmed cell death that is characterized by excessive iron accumulation, elevated lipid peroxides, and the overproduction of ROS. The maintenance of iron homeostasis is contingent upon the activity of the transferrin receptor (TfR), ferritin (Ft), and ferroportin (FPn). In the retina, iron accumulation and lipid peroxidation can contribute to the development of age-related macular degeneration (AMD). This phenomenon can be explained by the occurrence of the Fenton reaction, in which the interaction between divalent iron and hydrogen peroxide leads to the generation of highly reactive hydroxyl radicals. The hydroxyl radicals exhibit a propensity to attack proteins, lipids, nucleic acids, and carbohydrates, thereby instigating oxidative damage and promoting lipid peroxidation. Ultimately, these processes culminate in cell death and retinal degeneration. In this context, a comprehensive understanding of the exact mechanisms underlying ferroptosis may hold significant importance for developing therapeutic interventions. This review summarizes recent findings on iron metabolism, cellular ferroptosis, and lipid metabolism in the aging retina. We also introduce developments in the therapeutic strategies using iron chelating agents. Further refinements of these knowledges would deepen our comprehension of the pathophysiology of AMD and advance the clinical management of degenerative retinopathy. A comprehensive search strategy was employed to identify relevant studies on the role of ferroptosis in AMD. We performed systematic searches of the PubMed and Web of Science electronic databases from inception to the current date. The keywords used in the search included "ferroptosis", "AMD", "age-related macular degeneration", "iron metabolism", "oxidative stress", and "ferroptosis pathways". Peer-reviewed articles, including original research, reviews, meta-analyses, and clinical studies, were included in this paper, with a focus on the molecular mechanisms of ferroptosis in AMDs. Studies not directly related to ferroptosis, iron metabolism, or oxidative stress in the context of AMD were excluded. Furthermore, articles that lacked sufficient data or were not peer-reviewed (e.g., conference abstracts, editorials, or opinion pieces) were not considered.
Collapse
Affiliation(s)
- Na Zhao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Hao Wu
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Dong Wei
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Ning Pu
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Kexin Wang
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Yashuang Liu
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Ye Tao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Zongming Song
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| |
Collapse
|
9
|
Yuan D, Guo T, Zhu X, Song W, Nie D, Yu H. Icariside II induces ferroptosis through the down-regulation of SLC7A11 in ovarian cancer. J Ovarian Res 2025; 18:71. [PMID: 40186304 PMCID: PMC11971851 DOI: 10.1186/s13048-025-01650-1] [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: 07/24/2024] [Accepted: 03/16/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Ovarian cancer (OV) is the leading cause of death among gynecological malignancies. This study aimed to investigate the influence of Icariside II on OV in vitro and in vivo and to elucidate whether Icariside II induces ferroptosis in OV cells by regulating SLC7A11 expression. METHODS SKOV3 cells and OV nude mice were treated with Icariside II, a control-plasmid or an SLC7A11-plasmid. EdU assay, flow cytometry, wound-healing assay, and Transwell assays were used to assess cell proliferation, apoptosis, migration, and invasion respectively. Total iron, Fe2+ levels, and intracellular lipid reactive oxygen species (ROS) stimulation were evaluated in both cells and tissues. Levels of cysteine (Cys), glutathione (GSH), and glutathione peroxidase 4 (GPX4) were also analyzed. Ferroptosis markers, including Ptgs2, Chac1, SLC7A11, and apoptosis-associated genes (Bax and Bcl-2), were detected using qRT-PCR, western blotting, and immunohistochemistry (IHC). SLC7A11 expression in OV was explored using data from The Cancer Genome Atlas (TCGA), and validated with IHC staining. RESULTS Icariside II-induced ferroptosis in OV cells was confirmed by elevated Fe2+ and total iron levels, enhanced lipid ROS levels, higher Ptgs2 and Chac1 mRNA levels, and reduced levels of SLC7A11, Cys, GSH, and GPX4 in both in vitro and in vivo models. These effects were partially reversed by the SLC7A11-plasmid. Moreover, Icariside II suppressed SKOV3 cell proliferation, inhibited cells migration and invasion, and promoted apoptosis by downregulating SLC7A11 expression. Furthermore, we found that SLC7A11 expression was upregulated in OV tissues compared to adjacent non-tumor tissues. CONCLUSION Icariside II induces ferroptosis in OV by downregulating SLC7A11 expression in vitro and in vivo. Our study identified Icariside II as a promising therapeutic agent for the treatment of OV.
Collapse
Affiliation(s)
- Donglan Yuan
- Department of Obstetrics and Gynecology, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China
| | - Ting Guo
- Center for Molecular Medicine, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China
| | - Xiaotong Zhu
- Department of Obstetrics and Gynecology, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China
| | - Weiwei Song
- Department of Traditional Chinese Medicine, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China
| | - Dengyun Nie
- Nanjing University of Traditional Chinese Medicine, Nanjing, 210023, China
| | - Hong Yu
- Department of Pathology, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China.
- Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, No. 399 Hailing Road, Taizhou, 225300, China.
| |
Collapse
|
10
|
Liu Y, Zhang JT, Sun M, Song J, Sun HM, Wang MY, Wang CM, Liu W. Targeting ferroptosis in the treatment of ulcerative colitis by traditional Chinese medicine: A novel therapeutic strategies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156539. [PMID: 39987602 DOI: 10.1016/j.phymed.2025.156539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/04/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND The incidence of ulcerative colitis (UC) has been rising rapidly in recent years, and there is currently no effective method to prevent its recurrence. Owing to its long treatment duration, difficulty in treatment, prolonged remission, and high costs, it has attracted global attention. Exploring safe, effective, and sustainable treatment regimens has become an urgent global issue. Traditional Chinese medicine (TCM) has unique advantages such as low cost, low drug resistance, and fewer side effects, and has accumulated rich experience in the treatment of UC. PURPOSE Ferroptosis, as a new form of non-apoptotic cell death, is characterized by iron homeostatic imbalance and lipid peroxidation in the redox system. Studies have shown that inhibited ferroptosis in intestinal epithelial cells can protect the intestinal mucosa. Targeted intervention in ferroptosis may be a new direction for the treatment of UC. METHODS We conducted a systematic literature search with Google Scholar, PubMed, Web of Science, ScienceDirect and X-mol databases have been utilized to retrieve relevant literature up to October 2024, using keywords included ferroptosis, Inflammatory bowel disease (IBD), UC, Crohn's disease and TCM, Chinese traditional prescription, Chinese medicine extract and active ingredients. The existing literature was comprehensively studied and sorted out. RESULTS Currently, UC is mainly treated with drugs, including corticosteroids, amino salicylates, biologics, and immunomodulators, but drug resistance and adverse reactions are common. Increasing evidence suggests that TCM may treat UC by interfering with ferroptosis. Scholars have confirmed that TCM can inhibit ferroptosis, and recent studies have shown that TCM can not only inhibit iron dependent lipid peroxidation in intestinal cells but also enhance the antioxidant and anti-inflammatory abilities of intestinal mucosa, thus playing a role in the treatment of UC. This review explores the relevance of TCM intervention in ferroptosis and the treatment of UC, discusses the possible mechanisms of ferroptosis in UC, and aims to provide a basis for the diagnosis and treatment of UC. CONCLUSION It is revealed that TCM targeted ferroptosis has a good application prospect in the treatment of UC, providing a theoretical basis for elucidating the pathogenesis of UC and the study of TCM targeting ferroptosis regulating lipid metabolism in the treatment of UC, and providing a new perspective for the treatment of IBD in the future.
Collapse
Affiliation(s)
- Ying Liu
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Meng Sun
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Jian Song
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Hai-Ming Sun
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Meng-Yang Wang
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Chun-Mei Wang
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Wei Liu
- College of Pharmacy, Beihua University, jilin 132013, China.
| |
Collapse
|
11
|
Chen C, Wang H, Yang J, Zhao B, Lei Y, Li H, Yang K, Liu B, Diao Y. Sodium Iodate-Induced Ferroptosis in Photoreceptor-Derived 661W Cells Through the Depletion of GSH. Int J Mol Sci 2025; 26:2334. [PMID: 40076952 PMCID: PMC11900459 DOI: 10.3390/ijms26052334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Oxidative stress-induced photoreceptor cell death is closely associated with the etiology of age-related macular degeneration (AMD), and sodium iodate (SI) has been widely used as an oxidant stimulus in AMD models to induce retinal pigment epithelium (RPE) and photoreceptor cell death. However, the mechanism underlying SI-induced photoreceptor cell death remains controversial and unclear. In this study, we elucidate that ferroptosis is a critical form of cell death induced by SI in photoreceptor-derived 661W cells. SI disrupts system Xc-, leading to glutathione (GSH) depletion and triggering lipid peroxidation, thereby promoting ferroptosis in photoreceptor-derived 661W cells. Additionally, SI enhances intracellular Fe2+ levels, which further facilitates reactive oxygen species (ROS) accumulation, making the 661W cells more susceptible to ferroptosis. Exogenous GSH, as well as specific inhibitors of ferroptosis such as Fer-1 and antioxidants like NAC, significantly attenuate SI-induced ferroptosis in photoreceptor-derived 661W cells. These findings provide new insights into the mechanisms of ferroptosis as a key pathway in SI-induced photoreceptor-derived 661W cell death.
Collapse
Affiliation(s)
- Chao Chen
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China; (H.W.); (J.Y.); (Y.L.); (Y.D.)
| | - Han Wang
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China; (H.W.); (J.Y.); (Y.L.); (Y.D.)
| | - Jiuyu Yang
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China; (H.W.); (J.Y.); (Y.L.); (Y.D.)
| | - Bi Zhao
- Yunnan Key Laboratory of Tea Science, Tea Research Institute, Yunnan Academy of Agricultural Science, Kunming 650201, China;
| | - Yutian Lei
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China; (H.W.); (J.Y.); (Y.L.); (Y.D.)
| | - Hanqiao Li
- School of Medicine, Xiamen University, Xiamen 361000, China; (H.L.); (K.Y.)
| | - Kunhuan Yang
- School of Medicine, Xiamen University, Xiamen 361000, China; (H.L.); (K.Y.)
| | - Benying Liu
- Yunnan Key Laboratory of Tea Science, Tea Research Institute, Yunnan Academy of Agricultural Science, Kunming 650201, China;
| | - Yong Diao
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China; (H.W.); (J.Y.); (Y.L.); (Y.D.)
| |
Collapse
|
12
|
Wu P, Zhao L, Du Y, Lu J, He Y, Shu Q, Peng H, Wang X. Melatonin protects retinal pigment epithelium cells against ferroptosis in AMD via the PI3K/AKT/MDM2/P53 pathway. Front Pharmacol 2025; 16:1543575. [PMID: 40083383 PMCID: PMC11903707 DOI: 10.3389/fphar.2025.1543575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/21/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction Oxidative stress-prompted degeneration of the retinal pigment epithelium (RPE) notably contributes to the onset of age-related macular degeneration (AMD). However, the pathways leading to RPE deterioration and possible preventative strategies are not yet completely comprehended. Methods Ferroptosis was assayed through the evaluation of lipid peroxidation (C11-BODIPY and MDA), reactive oxygen species (ROS), transmission electron microscopy (TEM), iron content measurement, q-PCR, western blotting, and immunofluorescence. To assess the structure and retinal function of RPE in mice, ERG (electroretinography), OCT (optical coherence tomography), and H&E (hematoxylin and eosin) staining were employed. Network pharmacology methods were utilized to elucidate the potential mechanisms underlying melatonin's protective effects against ferroptosis in RPE cells in AMD. Genetic engineering techniques were applied to investigate the regulatory relationships among phosphatidylinositol 3-kinase (PI3K), protein kinase-B (AKT), murine double minute-2 (MDM2), protein 53 (P53), and solute carrier family 7 member 11 (SLC7A11). In vitro knockdown experiments of MDM2 were conducted to explore its regulatory role in ferroptosis within RPE cells. Results Aβ1-40 can trigger ferroptosis in RPE cells. Melatonin can inhibit the oxidative stress and ferroptosis induced by Aβ1-40 in RPE cells. Melatonin exhibits a protective effect on Aβ1-40-induced AMD, significantly improving the structure of the mouse retina and RPE layer, and facilitating the restoration of visual function. Network pharmacology methods revealed that the potential targets of melatonin in AMD are closely related to ferroptosis, and indicated that the predominant pathways are significantly associated with the PI3K/AKT/MDM2/P53 signaling pathway. Knocking down the specific expression of MDM2 can significantly weaken the inhibitory effect of melatonin on oxidative stress and ferroptosis. Discussion Melatonin can suppress cell death by ferroptosis in RPE via the PI3K/AKT/MDM2/P53 pathway, thereby preventing and decelerating the progression of AMD.
Collapse
Affiliation(s)
- Ping Wu
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ophthalmology, Chongqing Aier Eye Hospital, Chongqing, China
| | - Long Zhao
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Du
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Lu
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxia He
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinxin Shu
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Peng
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xing Wang
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Yang Y, Liu M, Dong X, Bai J, Shi W, Zhu Q, Liu J, Wang Z, Yi L, Yin X, Ni J, Qu C. Naringin Suppresses CoCl 2-Induced Ferroptosis in ARPE-19 Cells. Antioxidants (Basel) 2025; 14:236. [PMID: 40002420 PMCID: PMC11852185 DOI: 10.3390/antiox14020236] [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: 01/08/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Hypoxic damage to retinal pigment epithelial (RPE) cells and subsequent neovascularization are key factors in the pathogenesis of branch retinal vein occlusion (BRVO). Naringin (NG), a naturally occurring flavanone glycoside, has demonstrated significant antioxidant and anti-neovascular activities. However, the regulatory effects and mechanisms of NG on ferroptosis in BRVO are yet to be explored. Our study aimed to investigate the protective effects of NG on RPE cells under hypoxic stress and to elucidate the underlying molecular mechanisms. Our findings revealed that NG significantly reduced cytotoxicity induced by cobaltous chloride (CoCl2) and also inhibited vascular proliferation in the retina, thereby attenuating choroidal neovascularization. NG pretreatment largely countered the overproduction of reactive oxygen species (ROS) and malondialdehyde (MDA) triggered by hypoxic damage, while also restoring levels of the antioxidants glutathione (GSH) and superoxide dismutase (SOD). Furthermore, NG pretreatment significantly activated the expression of hypoxia-inducible factor-1 alpha (HIF-1α) and its downstream heme oxygenase-1 (HO-1) and NADPH dehydrogenase (NQO1). In conclusion, NG not only inhibits neovascularization but also alleviates inflammation in RPE cells by modulating the HO-1/GPX4 pathway to inhibit ferroptosis. These findings highlight the potential of NG as a promising therapeutic agent for the treatment of BRVO.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jian Ni
- College of Chinese Herbal Medicine, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.Y.); (M.L.); (X.D.); (J.B.); (Q.Z.); (J.L.); (Z.W.); (L.Y.); (X.Y.)
| | - Changhai Qu
- College of Chinese Herbal Medicine, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.Y.); (M.L.); (X.D.); (J.B.); (Q.Z.); (J.L.); (Z.W.); (L.Y.); (X.Y.)
| |
Collapse
|
14
|
Zhang A, Wei TT, Tan X, Tan CY, Zhuang M, Xie TH, Cai J, Yao Y, Zhu L. FADS1 inhibition protects retinal pigment epithelium cells from ferroptosis in age related macular degeneration. Eur J Pharmacol 2025; 989:177227. [PMID: 39736411 DOI: 10.1016/j.ejphar.2024.177227] [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: 09/10/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/01/2025]
Abstract
PURPOSE Age-related macular degeneration (AMD) is the leading cause of vision loss among the elderly individuals. Retinal pigment epithelium (RPE) ferroptosis is a significant pathogenetic component in AMD. This study aims to elucidate the role and mechanisms of fatty acid desaturase 1 (FADS1) in ferroptosis as well as AMD progression. METHODS An integrated bioinformatics analysis based on the array of data from the GEO database was conducted to identify candidates involved in ferroptosis during AMD. Subsequently, cellular and mouse models of AMD were developed using sodium iodate (NaIO3) to confirm the altered expression of FADS1. After treatment with a FADS1 inhibitor, cell survival, lipid peroxidation, and indicators of AMD were assessed in vitro and in vivo models. Further, immunofluorescence, immunohistochemistry, and swept-source OCT imaging were performed to assess the impacts of pharmacological inhibition of transcription factor specificity protein 1 (Sp1) on FADS1 and ferroptosis. RESULTS FADS1 expression was upregulated in AMD patients and in vitro and in vivo models of AMD. Its pharmacological inhibition had decreased mitochondrial ROS formation, lipid peroxidation, and ferroptosis as well as increased RPE cell function in ARPE-19 cells and C57BL/6J mouse models of AMD. Mechanistically, Sp1 was identified as a key transcription factor of FADS1. Moreover, Sp1 inhibition downregulated FADS1 expression consequently attenuating FADS1-mediated ferroptosis as well as AMD phenotypes. CONCLUSION For the first time, we demonstrated that Sp1 regulates FADS1-mediated ferroptosis in RPE cells. Our findings provide novel insights into the progression and treatment of AMD.
Collapse
Affiliation(s)
- Ao Zhang
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Ting-Ting Wei
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Xin Tan
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Cheng-Ye Tan
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Miao Zhuang
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Tian-Hua Xie
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Jiping Cai
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Yong Yao
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| | - Lingpeng Zhu
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
15
|
Chen J, Zhan Q, Li L, Xi S, Cai L, Liu R, Chen L. Cell-membrane targeting sonodynamic therapy combination with FSP1 inhibition for ferroptosis-boosted immunotherapy. Mater Today Bio 2025; 30:101407. [PMID: 39811609 PMCID: PMC11732120 DOI: 10.1016/j.mtbio.2024.101407] [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: 10/27/2024] [Revised: 12/02/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Cell membrane targeting sonodynamic therapy could induce the accumulation of lipid peroxidation (LPO), drive ferroptosis, and further enhances immunogenic cell death (ICD) effects. However, ferroptosis is restrained by the ferroptosis suppressor protein 1 (FSP1) at the plasma membrane, which can catalyze the regeneration of ubiquinone (CoQ10) by using NAD(P)H to suppress the LPO accumulation. This work describes the construction of US-active nanoparticles (TiF NPs), which combinate cell-membrane targeting sonosensitizer TBT-CQi with FSP1 inhibitor (iFSP1), facilitating cell-membrane targeting sonodynamic-triggered ferroptosis. TiF NPs could induce a sonodynamic effect, which promotes lipid peroxidation and drives apoptosis. Furthermore, TiF NPs could suppress FSP1, induce CoQ10 depletion, down-regulate the NADH, enhance LPO accumulation, and finally induce ferroptosis. In vitro results demonstrated that synergetic cell membrane targeting SDT/FSP1 inhibition triggered immunogenic cell death (ICD). Moreover, the as-synthesized TiF NPs-mediated cell membrane targeting SDT/FSP1 inhibition thoroughly inhibited the tumor growth and simultaneously activated antitumor immunity to suppress lung metastasis. This work represents a promising tumor therapeutic strategy combining cell membrane targeting SDT and FSP1 inhibition, potentially inspiring further research in developing logical and effective cancer therapies based on synergistic SDT/ferroptosis.
Collapse
Affiliation(s)
- Jian Chen
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Qiyu Zhan
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Lie Li
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Simin Xi
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Longmei Cai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Ruiyuan Liu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Lujia Chen
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| |
Collapse
|
16
|
Tian W, Su X, Hu C, Chen D, Li P. Ferroptosis in thyroid cancer: mechanisms, current status, and treatment. Front Oncol 2025; 15:1495617. [PMID: 39917169 PMCID: PMC11798778 DOI: 10.3389/fonc.2025.1495617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Thyroid cancer (TC) represents the most prevalent malignancy within the endocrine system. In recent years, there has been a marked global increase in the incidence of thyroid cancer, garnering substantial scientific interest. Comprehensive investigations into the pathogenesis of TC have identified a significant association with ferroptosis, a newly characterized form of cell death mediated by iron ions. Distinct from apoptosis, necrosis, and autophagy, ferroptosis is characterized by the accumulation of lipid peroxides and reactive oxygen species, culminating in cellular damage and death.Recent research has elucidated a connection between ferroptosis and the initiation, progression, and treatment of thyroid cancer. These findings underscore the significance of ferroptosis in thyroid cancer and offer valuable insights into the development of novel therapeutic strategies and precise predictive markers. The unique mechanisms of ferroptosis present opportunities for targeting treatment-resistant thyroid cancers. Consequently, the regulation of ferroptosis may emerge as a novel therapeutic target, potentially addressing the limitations of current treatments. Moreover, elucidating the molecular mechanisms underpinning ferroptosis in thyroid cancer may facilitate the identification of novel biomarkers for early detection and prognostication. This review endeavors to synthesize the extant knowledge regarding the role of ferroptosis in thyroid cancer, examine potential therapeutic implications, and propose future research trajectories to enhance the understanding and clinical application of ferroptosis.
Collapse
Affiliation(s)
- Wenzhi Tian
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Peking University-The Hong Kong University of Science and Technology Medical Centre, Shenzhen, Guangdong, China
- Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, China
| | - Xi Su
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Peking University-The Hong Kong University of Science and Technology Medical Centre, Shenzhen, Guangdong, China
| | - Chenchen Hu
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Peking University-The Hong Kong University of Science and Technology Medical Centre, Shenzhen, Guangdong, China
| | - Dong Chen
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Peking University-The Hong Kong University of Science and Technology Medical Centre, Shenzhen, Guangdong, China
| | - Peng Li
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Peking University-The Hong Kong University of Science and Technology Medical Centre, Shenzhen, Guangdong, China
| |
Collapse
|
17
|
Tao L, Xu J, Jiang L, Hu J, Tang Z. Investigation into the influence of mild hypothermia on regulating ferroptosis through the P53-SLC7A11/GPX4 signaling pathway in sepsis-induced acute lung injury. Intensive Care Med Exp 2025; 13:4. [PMID: 39812923 PMCID: PMC11735705 DOI: 10.1186/s40635-025-00713-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Sepsis-induced acute lung injury (S-ALI) significantly contributes to unfavorable clinical outcomes. Emerging evidence suggests a novel role for ferroptosis in the pathophysiology of ALI, though the precise mechanisms remain unclear. Mild hypothermia (32-34 °C) has been shown to inhibit inflammatory responses, reduce oxidative stress, and regulate metabolic processes. P53 has been reported to downregulate the transcriptional activity of solute carrier family 7 member 11 (SLC7A11), thereby limiting cystine uptake. This reduction in cystine availability compromises the activity of Glutathione peroxidase 4 (GPX4), a cystine-dependent enzyme, ultimately increasing cellular susceptibility to ferroptosis. However, it remains unclear whether mild hypothermia exerts protective effects through the P53-SLC7A11/GPX4 signaling pathway. This study investigates the influence of mild hypothermia on ferroptosis mediated by the P53-SLC7A11/GPX4 pathway in S-ALI. METHODS This study utilized both in vivo and in vitro models. In the vivo model, 64 Sprague-Dawley rats were employed, with 40 analyzed for survival outcomes. Sepsis was induced using the cecum ligation and puncture (CLP) method, after which rats were subjected to either normothermic (36-38 °C) or mild hypothermic (32-34 °C) conditions for a duration of 10 h. Twelve hours post-surgery, blood samples, bronchoalveolar lavage fluid, and lung tissue samples were harvested for histological analysis, measurement of inflammatory markers, wet/dry ratios, blood gas analysis, assessment of oxidative stress and ferroptosis, Western blotting, and RT-qPCR analysis. In the in vitro model, RLE-6TN cells were exposed to lipopolysaccharide (LPS) for 24 h under normothermic and mild hypothermic conditions. These cells were then evaluated for cell viability, inflammatory markers, oxidative stress levels, ferroptosis markers, as well as Western blot and RT-qPCR analyses. RESULTS CLP-induced sepsis led to elevated levels of inflammatory markers, increased lung injury scores, and heightened oxidative stress markers. These detrimental effects were significantly ameliorated by mild hypothermia. Furthermore, mild hypothermia reversed the modified expression of P53, SLC7A11, and GPX4 signaling molecules. Notably, mild hypothermia also improved the 5-day survival rate of CLP rats. CONCLUSION Mild hypothermia attenuates S-ALI and modulates ferroptosis through the P53-SLC7A11/GPX4 signaling pathway.
Collapse
Affiliation(s)
- Liujun Tao
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Jie Xu
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Liangyan Jiang
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Juntao Hu
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Zhanhong Tang
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
18
|
Chen Q, Song JX, Zhang Z, An JR, Gou YJ, Tan M, Zhao Y. Exploring Liraglutide's mechanism in reducing renal fibrosis: the Fsp1-CoQ10-NAD(P)H pathway. Sci Rep 2025; 15:1754. [PMID: 39799153 PMCID: PMC11724886 DOI: 10.1038/s41598-025-85658-z] [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: 07/26/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
Studies have confirmed that elevated glucose levels could lead to renal fibrosis through the process of ferroptosis. Liraglutide, a human glucagon-like peptide-1 (GLP-1) analogue, is a potential treatment option for diabetes. This study aimed to examine the potential of liraglutide (LIRA) in inhibiting ferroptosis and reducing high glucose-induced renal fibrotic injury in mice, and whether the Fsp1-CoQ10-NAD(P)H signal pathway is a mechanism for this effect. In our study, we used db/db mice to simulate Type 2 diabetes mellitus (T2DM). The mice were intraperitoneally injected with LIRA (200 µg/kg/d) daily for 6 weeks. Renal function, pathologic changes, lipid peroxidation levels, iron levels, and ferroptosis were assessed. First, LIRA ameliorated renal dysfunction and fibrosis in db/db mice. Second, LIRA inhibited lipid peroxidation by up-regulating T-SOD, GSH-Px, and GSH activities as well as down-regulating the levels of 8-OHDG, MDA, LPO, 4-HNE, 12-Lox, and NOX4 in db/db mice. In addition, LIRA attenuated iron deposition by decreasing the expression of TfR1 and increasing the expression of FPN1. Meanwhile, LIRA reduced high levels of high glucose-induced cell viability decline and intracellular lipid peroxidation. Furthermore, LIRA inhibited ferroptosis by adjusting the Fsp1-CoQ10-NAD(P)H pathway in vivo and in vitro. These findings suggested that LIRA attenuated kidney fibrosis injury in db/db mice by inhibiting ferroptosis through the Fsp1-CoQ10-NAD(P)H pathway.
Collapse
Affiliation(s)
- Qi Chen
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, NO.3, Luqian Xingyuan Road, Shijiazhuang, 050200, Hebei Province, China
- College of Basic Medicine, Chengde Medical University, Chengde, 067000, China
| | - Ji-Xian Song
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, NO.3, Luqian Xingyuan Road, Shijiazhuang, 050200, Hebei Province, China
| | - Zhi Zhang
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, NO.3, Luqian Xingyuan Road, Shijiazhuang, 050200, Hebei Province, China
| | - Ji-Ren An
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, NO.3, Luqian Xingyuan Road, Shijiazhuang, 050200, Hebei Province, China
| | - Yu-Jing Gou
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, NO.3, Luqian Xingyuan Road, Shijiazhuang, 050200, Hebei Province, China
- College of Basic Medicine, Chengde Medical University, Chengde, 067000, China
| | - Miao Tan
- The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| | - Yashuo Zhao
- Hebei Technology Innovation Center of TCM Combined Hydrogen Medicine, Hebei University of Chinese Medicine, NO.3, Luqian Xingyuan Road, Shijiazhuang, 050200, Hebei Province, China.
| |
Collapse
|
19
|
Ye Z, Yan Y, Jin F, Jiang J, Deng C, Wang L, Dong K. Deferiprone protects photoreceptors by inhibiting ferroptosis after experimental retinal detachment. Exp Eye Res 2025; 250:110156. [PMID: 39549870 DOI: 10.1016/j.exer.2024.110156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/26/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024]
Abstract
The detachment of the retinal neuroepithelium from the retinal pigment epithelium (RPE), often due to a retinal tear and subsequent subretinal fluid (SRF) accumulation, is a critical factor leading to photoreceptor cells (PR) death and permanent vision impairment in retinal detachment (RD) scenarios. Predicting postoperative visual recovery is challenging, even with surgical reattachment. Research has indicated that increased iron and transferrin (TF) saturation in the vitreous fluid (VF) correlates with poorer visual outcomes, suggesting a potential role for ferroptosis, a form of regulated cell death, in PR following RD. To explore this hypothesis, we analyzed the VF of RD patients for ferroptosis markers, revealing reduced levels of glutathione peroxidase 4 (GPX4), glutathione (GSH), and reduced nicotinamide adenine dinucleotide phosphate (NADPH), alongside elevated levels of Long-chain acyl-CoA synthetase 4(ACSL4), malondialdehyde (MDA), and ferrous iron. We then developed a mouse model to simulate RD and administered the iron chelator deferiprone (DFP) as a treatment. Our findings indicated that DFP mitigated ferroptosis in the retina, thereby preserving retinal architecture and function. Collectively, our study establishes the occurrence of ferroptosis in RD and demonstrates the therapeutic potential of DFP in protecting PR and treating RD.
Collapse
Affiliation(s)
- Ziyang Ye
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
| | - Yuanye Yan
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
| | - Feiyu Jin
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
| | - Jiazhen Jiang
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
| | - Can Deng
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China
| | - Lisong Wang
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China.
| | - Kai Dong
- Department of Ophthalmology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China.
| |
Collapse
|
20
|
Huang J, Zhao Y, Luo X, Luo Y, Ji J, Li J, Lai J, Liu Z, Chen Y, Lin Y, Liu J. Dexmedetomidine inhibits ferroptosis and attenuates sepsis-induced acute kidney injury via activating the Nrf2/SLC7A11/FSP1/CoQ10 pathway. Redox Rep 2024; 29:2430929. [PMID: 39581576 PMCID: PMC11587839 DOI: 10.1080/13510002.2024.2430929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024] Open
Abstract
OBJECTIVES The molecular mechanism underlying the protective effects of DEX against sepsis-induced acute kidney injury (SAKI) remains to be elucidated. METHODS We established S-AKI models in vivo via CLP and in vitro with LPS-induced HK-2 cells. RESULTS The Nrf2/SLC7A11/FSP1/CoQ10 pathway was inhibited in S-AKI both in vitro and in vivo. The overexpression of Nrf2 inhibited LPS-induced ferroptosis by activating the SLC7A11/FSP1/CoQ10 pathway. DEX ameliorated kidney tissue damage, as determined by a decrease in BUN, Cr, and inflammatory factor levels, along with renal tubule vacuolation and inflammatory cell infiltration in S-AKI mice. Additionally, DEX treatment significantly ameliorated ferroptosis in S-AKI in vitro and in vivo, as indicated by an improvement in mitochondrial shrinkage and disruption of cristae, a decrease in iron, ROS, MDA, and 4-HNE levels, and an increase in GSH and GPX4 levels. Mechanistically, DEX treatment restored the reduction of Nrf2 expression and nuclear translocation in S-AKI, as well as, the levels of downstream SLC7A11, FSP1, and CoQ10. Knocking down Nrf2 in vitro and administering brusatol in vivo eliminated the protective effect of DEX against S-AKI. CONCLUSIONS DEX mitigated ferroptosis and attenuated S-AKI by activating the Nrf2/SLC7A11/FSP1/CoQ10 pathway. Abbreviation: CLP: Cecal ligation puncture; LPS: Lipopolysaccharide; Nrf2: Nuclear factor-erythroid- 2-related factor 2; SLC7A11: Solute carrier family 7 member 11; FSP1: Ferroptosis suppressor protein 1; CoQ10: Coenzyme Q10; BUN: Blood urea nitrogen; Cr: Serum creatinine; ROS: Reactive oxygen species; MDA: Malondialdehyde; 4-HNE: 4-hydroxynonenal; GSH: Hlutathione; GPX4: Glutathione peroxidase 4.
Collapse
Affiliation(s)
- Jiao Huang
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Yang Zhao
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, People’s Republic of China
| | - Xi Luo
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Yunpeng Luo
- Department of Anesthesiology, Guizhou Provincial People’s Hospital, Guiyang, People’s Republic of China
| | - Jiemei Ji
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Jia Li
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Jian Lai
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Ziru Liu
- Department of Anesthesiology, Yueyang Central Hospital, Yueyang, People’s Republic of China
| | - Yuanyuan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Yunan Lin
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Jingchen Liu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| |
Collapse
|
21
|
He D, Tan XN, Li LP, Gao WH, Tian XF, Zeng PH. Brazilin Actuates Ferroptosis in Breast Cancer Cells via p53/SLC7A11/GPX4 Signaling Pathway. Chin J Integr Med 2024; 30:1001-1006. [PMID: 38652227 DOI: 10.1007/s11655-024-4104-y] [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] [Accepted: 01/03/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVE To investigate the mechanism of induction of ferroptosis by brazilin in breast cancer cells. METHODS Breast cancer 4T1 cells were divided into 6 groups: control, brazilin 1/2 half maximal inhibitory concentration (IC50), IC50, 2×IC50, erastin (10 µg/mL) and capecitabine (10 µg/mL) groups. The effect of brazilin on the proliferation of 4T1 cells was detected by cell counting kit-8 assay, and the treatment dose of brazilin was screened. The effect of brazilin on the mitochondrial morphology of 4T1 cells, and the mitochondrial damage was evaluated under electron microscopy. The levels of Fe2+, reactive oxygen species (ROS), malondialdehyde (MDA), glutathione (GSH) and glutathione peroxidase 4 (GPX4) were estimated using various detection kits. The invasion and migration abilities of 4T1 cells were detected by scratch assay and transwell assay. The expressions levels of tumor protein p53, solute carrier family 7 member 11 (SLC7A11), GPX4 and acyl-CoA synthetase long-chain family member 4 (ACSL4) proteins were quantified by Western blot assay. RESULTS Compared to the control group, the 10 (1/2 IC50), 20 (IC50) and 40 (2×IC50) µg/mL brazilin, erastin, and capecitabine groups showed a significant decrease in the cell survival rate, invasion and migration abilities, GSH, SLC7A11 and GPX4 protein expression levels, and mitochondrial volume and ridge (P<0.05), and a significant increase in the mitochondria membrane density, Fe2+, ROS and MDA levels, and p53 and ACSL4 protein expression levels (P<0.05). CONCLUSIONS Brazilin actuated ferroptosis in breast cancer cells, and the underlying mechanism is mainly associated with the p53/SLC7A11/GPX4 signaling pathway.
Collapse
Affiliation(s)
- Dan He
- Hunan Academy of Chinese Medicine, Changsha, 410006, China
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiao-Ning Tan
- Scientific Research Department, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, China
| | - Lin-Pei Li
- Oncology Department, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, China
| | - Wen-Hui Gao
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xue-Fei Tian
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Pu-Hua Zeng
- Hunan Academy of Chinese Medicine, Changsha, 410006, China.
| |
Collapse
|
22
|
Xia J, Li X, Bai C, Han X. Research Progress of Coenzyme Q in Diabetes Mellitus and Its Common Complications. Diabetes Metab Syndr Obes 2024; 17:3629-3641. [PMID: 39376660 PMCID: PMC11457790 DOI: 10.2147/dmso.s481690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
Coenzyme Q has garnered significant attention due to its potential role in enhancing cellular energy production and its antioxidant properties. We delve into the therapeutic potential of coenzyme Q in managing diabetes mellitus and its complications, highlighting its capacity to improve mitochondrial function, reduce inflammation and oxidative stress, and correct lipid profiles. Coenzyme Q has shown promise in ameliorating insulin resistance and alleviating complications such as diabetic peripheral neuropathy, kidney disease, retinopathy, and cardiomyopathy. However, its clinical application is limited by poor bio-availability. This review also provides a comprehensive overview of current therapeutic strategies for diabetes complications involving coenzyme Q, including stimulating endogenous synthesis and utilizing carrier transport systems, offering insights into mechanisms for enhancing coenzyme Q bio-availability. These findings suggest that, with improved delivery methods, coenzyme Q could become a valuable adjunct therapy in the management of diabetes mellitus.
Collapse
Affiliation(s)
- Jingdong Xia
- Affiliated Hospital of Chifeng University, Chifeng, The Inner Mongol Autonomous Region, Chifeng, People’s Republic of China
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng, People’s Republic of China
| | - Xiudan Li
- Affiliated Hospital of Chifeng University, Chifeng, The Inner Mongol Autonomous Region, Chifeng, People’s Republic of China
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng, People’s Republic of China
| | - Chunying Bai
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng, People’s Republic of China
| | - Xuchen Han
- Affiliated Hospital of Chifeng University, Chifeng, The Inner Mongol Autonomous Region, Chifeng, People’s Republic of China
| |
Collapse
|
23
|
Muluh TA, Fu Q, Ai X, Wang C, Chen W, Zheng X, Wang W, Wang M, Shu XS, Ying Y. Targeting Ferroptosis as an Advance Strategy in Cancer Therapy. Antioxid Redox Signal 2024; 41:616-636. [PMID: 38959114 DOI: 10.1089/ars.2024.0608] [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] [Indexed: 07/05/2024]
Abstract
Significance: This study innovates by systematically integrating the molecular mechanisms of iron death and its application in cancer therapy. By deeply analyzing the interaction between iron death and the tumor microenvironment, the study provides a new theoretical basis for cancer treatment and directions for developing more effective treatment strategies. In addition, the study points to critical issues and barriers that need to be addressed in future research, providing valuable insights into the use of iron death in clinical translation. Recent Advances: These findings are expected to drive further advances in cancer treatment, bringing patients more treatment options and hope. Through this paper, we see the great potential of iron death in cancer treatment and look forward to more research results being translated into clinical applications in the future to contribute to the fight against cancer. Critical Issues: In today's society, cancer is still one of the major diseases threatening human health. Despite advances in existing treatments, cancer recurrence and drug resistance remain a severe problem. These problems increase the difficulty of treatment and bring a substantial physical and mental burden to patients. Therefore, finding new treatment strategies to overcome these challenges has become significant. Future Directions: The study delved into the molecular basis of iron death in tumor biology. It proposed a conceptual framework to account for the interaction of iron death with the tumor immune microenvironment, guide treatment selection, predict efficacy, explore combination therapies, and identify new therapeutic targets to overcome cancer resistance to standard treatments, peeving a path for future research and clinical translation of ferroptosis as a potential strategy in cancer therapy. Antioxid. Redox Signal. 41, 616-636. [Figure: see text].
Collapse
Affiliation(s)
- Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Qianqian Fu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiaojiao Ai
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Changfeng Wang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Wei Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiangyi Zheng
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Wei Wang
- Shanghai Waker Bioscience Co., Ltd., Shanghai, China
| | - Maolin Wang
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xing-Sheng Shu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Ying Ying
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| |
Collapse
|
24
|
Xue P, Zhuang H, Shao S, Bai T, Zeng X, Yan S. Engineering Biodegradable Hollow Silica/Iron Composite Nanozymes for Breast Tumor Treatment through Activation of the "Ferroptosis Storm". ACS NANO 2024; 18:25795-25812. [PMID: 39226614 DOI: 10.1021/acsnano.4c08574] [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: 09/05/2024]
Abstract
The activation of cellular ferroptosis is promising in tumor therapy. However, ferroptosis is parallelly inhibited by antiferroptotic substances, including glutathione peroxidase 4 (GPX4), dihydroorotate dehydrogenase (DHODH), and ferroptosis suppressor protein 1 (FSP1). Thus, it is highly desirable, yet challenging, to simultaneously suppress these three antiferroptotic substances for activating ferroptosis. Here, we rationally designed a hollow iron-doped SiO2-based nanozyme (FeSHS) loaded with brequinar (BQR) and lificiguat (YC-1), named FeSHS/BQR/YC-1-PEG, for tumor ferroptosis activation. FeSHS were developed through the continuous etching of SiO2 nanoparticles by iron ions, which exhibit pH/glutathione-responsive biodegradability, along with mimicking the activities of peroxidase, glutathione oxidase, and NAD(P)H oxidase. Specifically, glutathione depletion and NAD(P)H oxidation by FeSHS will suppress the expression of GPX4 and inhibit FSP1 by disrupting the NAD(P)H/FSP1/ubiquinone axis. In addition, the released BQR can suppress the expression of DHODH. Meanwhile, YC-1 is able to increase the cellular polyunsaturated fatty acids (PUFAs) by destroying the HIF-1α/lipid droplet axis. The elevation of levels of iron and PUFAs while simultaneously disrupting the GPX4/DHODH/FSP1 inhibitory pathways by our designed nanoplatform displayed high therapeutic efficacy both in vitro and in vivo. This work elucidates rationally designing smart nanoplatforms for ferroptosis activation and future tumor treatments.
Collapse
Affiliation(s)
- Panpan Xue
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, China
| | - Huilan Zhuang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, China
| | - Sijie Shao
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, China
| | - Tingjie Bai
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, China
| | - Xuemei Zeng
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, 1 Keji Road, Fuzhou 350117, PR China
| | - Shuangqian Yan
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, China
| |
Collapse
|
25
|
Zhu H, Yang Y, Duan Y, Zheng X, Lin Z, Zhou J. Nrf2/FSP1/CoQ10 axis-mediated ferroptosis is involved in sodium aescinate-induced nephrotoxicity. Arch Biochem Biophys 2024; 759:110100. [PMID: 39033970 DOI: 10.1016/j.abb.2024.110100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Sodium aescinate (SA), an active compound found in horse chestnut seeds, is widely used in clinical practice. Recently, the incidence of SA-induced adverse events, particularly renal impairment, has increased. Our previous work demonstrated that SA causes severe nephrotoxicity via nephrocyte ferroptosis; however, the underlying mechanism remains to be fully elucidated. In the current study, we investigated additional molecular pathways involved in SA-induced nephrotoxicity. Our results showed that SA inhibited cell viability, disrupted cellular membrane integrity, and enhanced reactive oxygen species (ROS), ferrous iron (Fe2+), and malondialdehyde (MDA) levels, as well as lipid peroxidation in rat proximal renal tubular epithelial cell line (NRK-52E) cells. SA also depleted coenzyme Q10 (CoQ10, ubiquinone) and nicotinamide adenine dinucleotide (NADH) and reduced ferroptosis suppressor protein 1 (FSP1) and polyprenyltransferase (coenzyme Q2, COQ2) activity, triggering lipid peroxidation and ROS accumulation in mouse kidneys and NRK-52E cells. The overexpression of COQ2, FSP1, or CoQ10 (ubiquinone) supplementation effectively attenuated SA-induced ferroptosis, whereas iFSP1 or 4-formylbenzoic acid (4-CBA) pretreatment exacerbated SA-induced nephrotoxicity. Additionally, SA decreased nuclear factor-erythroid-2-related factor 2 (Nrf2) levels and inhibited Nrf2 binding to the -1170/-1180 bp ARE site in FSP1 promoter, resulting in FSP1 suppression. Overexpression of Nrf2 or its agonist dimethyl fumarate (DMF) promoted FSP1 expression, thereby improving cellular antioxidant capacity and alleviating SA-induced ferroptosis. These results suggest that SA-triggers renal injury through oxidative stress and ferroptosis, driven by the suppression of the Nrf2/FSP1/CoQ10 axis.
Collapse
Affiliation(s)
- Haiyan Zhu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Yijing Yang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Yenan Duan
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Xin Zheng
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Zixiong Lin
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China.
| |
Collapse
|
26
|
Huang K, Deng H, Wang S, Zhang F, Huang G, Wang L, Liu J, Zhao X, Ren H, Yang G, Lin Z. Melanin-Like Nanomedicine Functions as a Novel RPE Ferroptosis Inhibitor to Ameliorate Retinal Degeneration and Visual Impairment in Dry Age-Related Macular Degeneration. Adv Healthc Mater 2024:e2401613. [PMID: 39129350 DOI: 10.1002/adhm.202401613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/01/2024] [Indexed: 08/13/2024]
Abstract
Ferrous ion accumulation and lethal oxidative stress mediate irreversible retinal pigment epithelial (RPE) cell ferroptosis and subsequent photoreceptor degeneration, a potential key pathogenic factor in the onset of dry age-related macular degeneration (dAMD), causing irreversible vision loss in the global elderly population. However, currently, no effective interventional treatment strategy exists in clinical practice. Herein, lesion site-targeted melanin-like nanoparticles, named ConA-MelNPs, are designed as a novel ferroptosis inhibitor for retinal degenerative diseases. ConA-MelNPs possessed chelating iron ion characteristics, alleviating severe mitochondrial damage caused by oxidative stress and protecting RPE cells from ferroptosis induced by sodium iodate (NaIO3). In a preclinical dAMD mouse model, a single intravitreal injection of ConA-MelNPs yielded significant responses in electroretinograms and visually-driven optomotor responses in visually impaired mice, resisting the challenge posed by secondary NaIO3-induced injuries, with the long-term sustainability of its therapeutic effect. Mechanistically, ConA-MelNPs achieve a therapeutic effect by interrupting the detrimental cascade involving "RPE cell ferroptosis, lethal oxidative stress, and microglial proinflammatory activation," affording the restoration of retinal homeostasis. The synthesized ConA-MelNPs demonstrated good biosafety, with no detected ophthalmic or systemic side effects. Collectively, ConA-MelNPs are proposed as a promising therapeutic option for atrophic retinal diseases such as dAMD.
Collapse
Affiliation(s)
- Keke Huang
- Department of Ophthalmology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Haoyue Deng
- Department of Anaesthesiology, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shuang Wang
- Department of Ophthalmology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Fuxiao Zhang
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Ge Huang
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Lu Wang
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Jianyu Liu
- Department of Neurology, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Xuli Zhao
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Hui Ren
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Guang Yang
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Zhiqing Lin
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| |
Collapse
|
27
|
Dai Q, Wei X, Zhao J, Zhang D, Luo Y, Yang Y, Xiang Y, Liu X. Inhibition of FSP1: A new strategy for the treatment of tumors (Review). Oncol Rep 2024; 52:105. [PMID: 38940330 PMCID: PMC11228423 DOI: 10.3892/or.2024.8764] [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: 02/23/2024] [Accepted: 05/10/2024] [Indexed: 06/29/2024] Open
Abstract
Ferroptosis, a regulated form of cell death, is intricately linked to iron‑dependent lipid peroxidation. Recent evidence strongly supports the induction of ferroptosis as a promising strategy for treating cancers resistant to conventional therapies. A key player in ferroptosis regulation is ferroptosis suppressor protein 1 (FSP1), which promotes cancer cell resistance by promoting the production of the antioxidant form of coenzyme Q10. Of note, FSP1 confers resistance to ferroptosis independently of the glutathione (GSH) and glutathione peroxidase‑4 pathway. Therefore, targeting FSP1 to weaken its inhibition of ferroptosis may be a viable strategy for treating refractory cancer. This review aims to clarify the molecular mechanisms underlying ferroptosis, the specific pathway by which FSP1 suppresses ferroptosis and the effect of FSP1 inhibitors on cancer cells.
Collapse
Affiliation(s)
- Qiangfang Dai
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Xiaoli Wei
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Jumei Zhao
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Die Zhang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yidan Luo
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yue Yang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yang Xiang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
- College of Physical Education, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Xiaolong Liu
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| |
Collapse
|
28
|
Chen Y, Pan G, Wu F, Zhang Y, Li Y, Luo D. Ferroptosis in thyroid cancer: Potential mechanisms, effective therapeutic targets and predictive biomarker. Biomed Pharmacother 2024; 177:116971. [PMID: 38901201 DOI: 10.1016/j.biopha.2024.116971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/28/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
Thyroid cancer is a prevalent endocrine malignancy whose global incidence has risen over the past several decades. Ferroptosis, a regulated form of cell death distinguished by the excessive buildup of iron-dependent lipid peroxidates, stands out from other programmed cell death pathways in terms of morphological and molecular characteristics. Increasing evidence suggests a close association between thyroid cancer and ferroptosis, that is, inducing ferroptosis effectively suppresses the proliferation of thyroid cancer cells and impede tumor advancement. Therefore, ferroptosis represents a promising therapeutic target for the clinical management of thyroid cancer in clinical settings. Alterations in ferroptosis-related genes hold potential for prognostic prediction in thyroid cancer. This review summarizes current studies on the role of ferroptosis in thyroid cancer, elucidating its mechanisms, therapeutic targets, and predictive biomarkers. The findings underscore the significance of ferroptosis in thyroid cancer and offer valuable insights into the development of innovative treatment strategies and accurate predictors for the thyroid cancer.
Collapse
Affiliation(s)
- Yuying Chen
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Gang Pan
- Department of Surgical Oncology, Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Fan Wu
- Department of Surgical Oncology, Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Yu Zhang
- Department of Surgical Oncology, Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Yuanhui Li
- Department of Surgical Oncology, Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, China.
| | - Dingcun Luo
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Department of Surgical Oncology, Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
29
|
You W, Azuma K, Iwagawa T, Watanabe S, Aihara M, Shiraya T, Ueta T. The role of lipid peroxidation in epithelial-mesenchymal transition of retinal pigment epithelial cells. Sci Rep 2024; 14:16498. [PMID: 39020017 PMCID: PMC11255318 DOI: 10.1038/s41598-024-67587-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024] Open
Abstract
Epithelial-Mesenchymal Transition (EMT) of retinal pigment epithelial (RPE) cells is recognized as pivotal in various retinal diseases. Previous studies have suggested a reciprocal regulation between reactive oxygen species (ROS) and EMT, though the involvement of peroxidized lipids or the effects of reducing them has remained unclear. The present study disclosed that EMT of ARPE-19 cells induced by TGF-β2 and TNF-α involves increased lipid peroxidation, and Ferrostatin-1 (Fer-1), a lipophilic antioxidative agent, successfully inhibited the increase in lipid peroxidation. Fer-1 suppressed the formation of EMT-associated fibrotic deposits, while EMT induction or Fer-1 treatment did not influence the cell viability or proliferation. Functionally, Fer-1 impeded EMT-driven cell migration and reduction in transepithelial electrical resistance. It demonstrated regulatory prowess by downregulating the mesenchymal marker fibronectin, upregulating the epithelial marker ZO-1, and inhibiting the EMT-associated transcriptional factor ZEB1. Additionally, VEGF, a major pathogenic cytokine in various retinal diseases, is also upregulated during EMT, and Fer-1 significantly mitigated the effect. The present study disclosed the involvement of lipid peroxidation in EMT of RPE cells, and suggests the suppression of lipid peroxidation may be a potential therapeutic target in retinal diseases in which EMT is implicated.
Collapse
Affiliation(s)
- Wang You
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Kunihiro Azuma
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Toshiro Iwagawa
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
- Department of Retinal Biology and Pathology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Sumiko Watanabe
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
- Department of Retinal Biology and Pathology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Tomoyasu Shiraya
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Takashi Ueta
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan.
| |
Collapse
|
30
|
Tan X, He Y, Yu P, Deng Y, Xie Z, Guo J, Hou Q, Li P, Lin X, Ouyang S, Ma W, Xie Y, Guo Z, Chen D, Zhang Z, Zhu Y, Huang F, Zhao Z, Zhang C, Guo Z, Chen X, Peng T, Li L, Xie W. The dual role of FSP1 in programmed cell death: resisting ferroptosis in the cell membrane and promoting necroptosis in the nucleus of THP-1 cells. Mol Med 2024; 30:102. [PMID: 39009982 PMCID: PMC11247902 DOI: 10.1186/s10020-024-00861-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/10/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Acute monocytic leukemia-M5 (AML-M5) remains a challenging disease due to its high morbidity and poor prognosis. In addition to the evidence mentioned earlier, several studies have shown that programmed cell death (PCD) serves a critical function in treatment of AML-M5. However, the role and relationship between ferroptosis and necroptosis in AML-M5 remains unclear. METHODS THP-1 cells were mainly treated with Erastin and IMP-366. The changes of ferroptosis and necroptosis levels were detected by CCK-8, western blot, quantitative real-time PCR, and electron microscopy. Flow cytometry was applied to detect the ROS and lipid ROS levels. MDA, 4-HNE, GSH and GSSG were assessed by ELISA kits. Intracellular distribution of FSP1 was studied by immunofluorescent staining and western blot. RESULTS The addition of the myristoylation inhibitor IMP-366 to erastin-treated acute monocytic leukemia cell line THP-1 cell not only resulted in greater susceptibility to ferroptosis characterized by lipid peroxidation, glutathione (GSH) depletion and mitochondrial shrinkage, as the FSP1 position on membrane was inhibited, but also increased p-RIPK1 and p-MLKL protein expression, as well as a decrease in caspase-8 expression, and triggered the characteristic necroptosis phenomena, including cytoplasmic translucency, mitochondrial swelling, membranous fractures by FSP1 migration into the nucleus via binding importin α2. It is interesting to note that ferroptosis inhibitor fer-1 reversed necroptosis. CONCLUSION We demonstrated that inhibition of myristoylation by IMP-366 is capable of switching ferroptosis and ferroptosis-dependent necroptosis in THP-1 cells. In these findings, FSP1-mediated ferroptosis and necroptosis are described as alternative mechanisms of PCD of THP-1 cells, providing potential therapeutic strategies and targets for AML-M5.
Collapse
Affiliation(s)
- Xiaoqian Tan
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan, China
| | - Yinling He
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Panpan Yu
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yunong Deng
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhongcheng Xie
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiami Guo
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Qin Hou
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pin Li
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyan Lin
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Ouyang
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wentao Ma
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yushu Xie
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Zilong Guo
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Dandan Chen
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Zhixia Zhang
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Yunyu Zhu
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Fei Huang
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Ziye Zhao
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Cen Zhang
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Zhirong Guo
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Xi Chen
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tianhong Peng
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Liang Li
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wei Xie
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
31
|
Lian J, Chen Y, Zhang Y, Guo S, Wang H. The role of hydrogen sulfide regulation of ferroptosis in different diseases. Apoptosis 2024:10.1007/s10495-024-01992-z. [PMID: 38980600 DOI: 10.1007/s10495-024-01992-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2024] [Indexed: 07/10/2024]
Abstract
Ferroptosis is a programmed cell death that relies on iron and lipid peroxidation. It differs from other forms of programmed cell death such as necrosis, apoptosis and autophagy. More and more evidence indicates that ferroptosis participates in many types of diseases, such as neurodegenerative diseases, ischemia-reperfusion injury, cardiovascular diseases and so on. Hence, clarifying the role and mechanism of ferroptosis in diseases is of great significance for further understanding the pathogenesis and treatment of some diseases. Hydrogen sulfide (H2S) is a colorless and flammable gas with the smell of rotten eggs. Many years ago, H2S was considered as a toxic gas. however, in recent years, increasing evidence indicates that it is the third important gas signaling molecule after nitric oxide and carbon monoxide. H2S has various physiological and pathological functions such as antioxidant stress, anti-inflammatory, anti-apoptotic and anti-tumor, and can participate in various diseases. It has been reported that H2S regulation of ferroptosis plays an important role in many types of diseases, however, the related mechanisms are not fully clear. In this review, we reviewed the recent literature about the role of H2S regulation of ferroptosis in diseases, and analyzed the relevant mechanisms, hoping to provide references for future in-depth researches.
Collapse
Affiliation(s)
- Jingwen Lian
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yuhang Chen
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yanting Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Shiyun Guo
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
32
|
Fan K, Wang J, Zhu W, Zhang X, Deng F, Zhang Y, Zou S, Kong L, Shi H, Li Z, Shen G, Wang D, Wu Z, Li H, Xu Z. Urinary proteomics for noninvasive monitoring of biomarkers of chronic mountain sickness in a young adult population using data-independent acquisition (DIA)-based mass spectrometry. J Proteomics 2024; 302:105195. [PMID: 38734407 DOI: 10.1016/j.jprot.2024.105195] [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: 03/18/2024] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Different populations exhibit varying pathophysiological responses to plateau environments. Therefore, it is crucial to identify molecular markers in body fluids with high specificity and sensitivity to aid in determination. Proteomics offers a fresh perspective for investigating protein changes linked to diseases. We utilize urine as a specific biomarker for early chronic mountain sickness (CMS) detection, as it is a simple-to-collect biological fluid. We collected urine samples from three groups: plains health, plateau health and CMS. Using DIA's proteomic approach, we found differentially expressed proteins between these groups, which will be used as a basis for future studies to identify protein markers. Compared with the healthy plain population, 660 altering proteins were identified in plateau health, which performed the resistance to altitude response function by boosting substance metabolism and reducing immune stress function. Compared to the healthy plateau population, the CMS group had 140 different proteins identified, out of which 8 were potential biomarkers for CMS. Our study has suggested that CMS may be closely related to increased thyroid hormone levels, oxidative damage to the mitochondria, impaired cell detoxification function and inhibited hydrolase activity. SIGNIFICANCE: Our team has compiled a comprehensive dataset of urine proteomics for AMS disease. We successfully identified differentially expressed proteins between healthy and AMS groups using the DIA proteomic approach. We discovered that 660 proteins were altered in plateau health compared to the healthy plain population, resulting in a heightened resistance to altitude response function by boosting substance metabolism and reducing immune stress function. Additionally, we pinpointed 140 different proteins in the AMS group compared to the healthy plateau population, with 8 showing potential as biomarkers for AMS. Our findings suggest that the onset of AMS may be closely linked to increased thyroid hormone levels, oxidative damage to the mitochondria, impaired cell detoxification function and inhibited hydrolase activity.
Collapse
Affiliation(s)
- Kaiyuan Fan
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China
| | - Jin Wang
- Department of Clinical Laboratory, Tianjin Third Central Hospital, Tianjin 300170, PR China
| | - Wenqing Zhu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China
| | - Xinan Zhang
- Xizang Corps Hospital of Chinese People's Armed Police Force, Lasa 850000, PR China
| | - Feng Deng
- Xizang Corps Hospital of Chinese People's Armed Police Force, Lasa 850000, PR China
| | - Yan Zhang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China
| | - Shuang Zou
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China
| | - Lingjia Kong
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China
| | - He Shi
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China
| | - Ziling Li
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China
| | - Guozheng Shen
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China
| | - Dong Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China
| | - Zhidong Wu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China.
| | - Heng Li
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China.
| | - Zhongwei Xu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, PR China; Tianjin key laboratory for prevention and control of occupational and environmental hazards, Tianjin 300309, PR China.
| |
Collapse
|
33
|
Yang Y, Lin Y, Han Z, Wang B, Zheng W, Wei L. Ferroptosis: a novel mechanism of cell death in ophthalmic conditions. Front Immunol 2024; 15:1440309. [PMID: 38994366 PMCID: PMC11236620 DOI: 10.3389/fimmu.2024.1440309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
Ferroptosis, a new type of programmed cell death proposed in recent years, is characterized mainly by reactive oxygen species and iron-mediated lipid peroxidation and differs from programmed cell death, such as apoptosis, necrosis, and autophagy. Ferroptosis is associated with a variety of physiological and pathophysiological processes. Recent studies have shown that ferroptosis can aggravate or reduce the occurrence and development of diseases by targeting metabolic pathways and signaling pathways in tumors, ischemic organ damage, and other degenerative diseases related to lipid peroxidation. Increasing evidence suggests that ferroptosis is closely linked to the onset and progression of various ophthalmic conditions, including corneal injury, glaucoma, age-related macular degeneration, diabetic retinopathy, retinal detachment, and retinoblastoma. Our review of the current research on ferroptosis in ophthalmic diseases reveals significant advancements in our understanding of the pathogenesis, aetiology, and treatment of these conditions.
Collapse
Affiliation(s)
- Yaqi Yang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yumeng Lin
- Naniing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
- Naniing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Bo Wang
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Wei Zheng
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Lijuan Wei
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
34
|
Mokhtarpour K, Razi S, Rezaei N. Ferroptosis as a promising targeted therapy for triple negative breast cancer. Breast Cancer Res Treat 2024:10.1007/s10549-024-07387-7. [PMID: 38874688 DOI: 10.1007/s10549-024-07387-7] [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: 12/06/2023] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE Triple negative breast cancer (TNBC) is a challenging subtype characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Standard treatment options are limited, and approximately 45% of patients develop distant metastasis. Ferroptosis, a regulated form of cell death triggered by iron-dependent lipid peroxidation and oxidative stress, has emerged as a potential targeted therapy for TNBC. METHODS This study utilizes a multifaceted approach to investigate the induction of ferroptosis as a therapeutic strategy for TNBC. It explores metabolic alterations, redox imbalance, and oncogenic signaling pathways to understand their roles in inducing ferroptosis, characterized by lipid peroxidation, reactive oxygen species (ROS) generation, and altered cellular morphology. Critical pathways such as Xc-/GSH/GPX4, ACSL4/LPCAT3, and nuclear factor erythroid 2-related factor 2 (NRF2) are examined for their regulatory roles in ferroptosis and their potential dysregulation contributing to cancer cell survival and resistance. RESULTS Inducing ferroptosis has been shown to inhibit tumor growth, enhance the efficacy of conventional therapies, and overcome drug resistance in TNBC. Lipophilic antioxidants, GPX4 inhibitors, and inhibitors of the Xc- system have been demonstrated to be potential ferroptosis inducers. Additionally, targeting the NRF2 pathway and exploring other ferroptosis regulators, such as ferroptosis suppressor protein 1 (FSP1), and the PERK-eIF2α-ATF4-CHOP pathway, may offer novel therapeutic avenues. CONCLUSION Further research is needed to understand the mechanisms, optimize therapeutic strategies, and evaluate the safety and efficacy of ferroptosis-targeted therapies in TNBC treatment. Overall, targeting ferroptosis represents a promising approach to improving treatment outcomes and overcoming the challenges posed by TNBC.
Collapse
Affiliation(s)
- Kasra Mokhtarpour
- Faculty of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Imunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
| | - Nima Rezaei
- Research Center for Imunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
35
|
Chen Z, Liu B, Zhou D, Lei M, Yang J, Hu Z, Duan W. AQP4 regulates ferroptosis and oxidative stress of Muller cells in diabetic retinopathy by regulating TRPV4. Exp Cell Res 2024; 439:114087. [PMID: 38735619 DOI: 10.1016/j.yexcr.2024.114087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
Diabetic retinopathy (DR) is a common microvascular complication that causes visual impairment or loss. Aquaporin 4 (AQP4) is a regulatory protein involved in water transport and metabolism. In previous studies, we found that AQP4 is related to hypoxia injury in Muller cells. Transient receptor potential cation channel subfamily V member 4 (TRPV4) is a non-selective cation channel protein involved in the regulation of a variety of ophthalmic diseases. However, the effects of AQP4 and TRPV4 on ferroptosis and oxidative stress in high glucose (HG)-treated Muller cells are unclear. In this study, we investigated the functions of AQP4 and TRPV4 in DR. HG was used to treat mouse Muller cells. Reverse transcription quantitative polymerase chain reaction was used to measure AQP4 mRNA expression. Western blotting was used to detect the protein levels of AQP4, PTGS2, GPX4, and TRPV4. Cell count kit-8, flow cytometry, 5,5',6,6'-tetrachloro-1,1,3,3'-tetraethylbenzimidazolyl carbocyanine iodide staining, and glutathione (GSH), superoxide dismutase (SOD), and malondialdehyde (MDA) kits were used to evaluate the function of the Muller cells. Streptozotocin was used to induce DR in rats. Haematoxylin and eosin staining was performed to stain the retina of rats. GSH, SOD, and MDA detection kits, immunofluorescence, and flow cytometry assays were performed to study the function of AQP4 and TRPV4 in DR rats. Results found that AQP4 and TRPV4 were overexpressed in HG-induced Muller cells and streptozotocin-induced DR rats. AQP4 inhibition promoted proliferation and cell cycle progression, repressed cell apoptosis, ferroptosis, and oxidative stress, and alleviated retinal injury in DR rats. Mechanistically, AQP4 positively regulated TRPV4 expression. Overexpression of TRPV4 enhanced ferroptosis and oxidative stress in HG-treated Muller cells, and inhibition of TRPV4 had a protective effect on DR-induced retinal injury in rats. In conclusion, inhibition of AQP4 inhibits the ferroptosis and oxidative stress in Muller cells by downregulating TRPV4, which may be a potential target for DR therapy.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China.
| | - Bingjie Liu
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| | - Daijiao Zhou
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Mingshu Lei
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Jingying Yang
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| | - Zhongyin Hu
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| | - Wenhua Duan
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| |
Collapse
|
36
|
Chen J, Duan Z, Deng L, Li L, Li Q, Qu J, Li X, Liu R. Cell Membrane-Targeting Type I/II Photodynamic Therapy Combination with FSP1 Inhibition for Ferroptosis-Enhanced Photodynamic Immunotherapy. Adv Healthc Mater 2024; 13:e2304436. [PMID: 38335308 DOI: 10.1002/adhm.202304436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/30/2024] [Indexed: 02/12/2024]
Abstract
An imbalance in reactive oxygen species (ROS) levels in tumor cells can result in the accumulation of lipid peroxide (LPO) which can induce ferroptosis. Moreover, elevated ROS levels in tumors present a chance to develop ROS-based cancer therapeutics including photodynamic therapy (PDT) and ferroptosis. However, their anticancer efficacies are compromised by insufficient oxygen levels and inherent cellular ROS regulatory mechanism. Herein, a cell membrane-targeting photosensitizer, TBzT-CNQi, which can generate 1O2, •OH, and O2 •- via type I/II process to induce a high level of LPO for potent ferroptosis and photodynamic therapy is developed. The FSP1 inhibitor (iFSP1) is incorporated with TBzT-CNQi to downregulate FSP1 expression, lower the intracellular CoQ10 content, induce a high level of LPO, and activate initial tumor immunogenic ferroptosis. In vitro and in vivo experiments demonstrate that the cell membrane-targeting type I/II PDT combination with FSP1 inhibition can evoke strong ICD and activate the immune response, which subsequently promotes the invasion of CD8+ T cells infiltration, facilitates the dendritic cell maturation, and decreases the tumor infiltration of tumor-associated macrophages. The study indicates that the combination of cell membrane-targeting type I/II PDT and FSP1 inhibition holds promise as a potential strategy for ferroptosis-enhanced photodynamic immunotherapy of hypoxia tumors.
Collapse
Affiliation(s)
- Jian Chen
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Zeyu Duan
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Lidong Deng
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Lie Li
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Qiyan Li
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jinqing Qu
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
| | - Xiang Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China
| | - Ruiyuan Liu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
37
|
Wang H, Zhao W, Wang D, Chen J. ANO6 (TMEM16F) inhibits gastrointestinal stromal tumor growth and induces ferroptosis. Open Med (Wars) 2024; 19:20240941. [PMID: 38756246 PMCID: PMC11097043 DOI: 10.1515/med-2024-0941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 05/18/2024] Open
Abstract
Herein, we elucidate the potential role of ANO6 (TMEM16F) in gastrointestinal stromal tumors (GISTs). ANO6 expression in GIST and adjacent normal tissues was determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting. Cell proliferation, apoptosis, and pyroptosis were examined utilizing 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, terminal deoxynucleotidyl transferase dUTP Nick-End Labeling staining, and flow cytometry. In addition, the total iron and Fe2+ levels were assessed. IL-18 and IL-1β levels were also evaluated. Lipid reactive oxygen species (ROS), cystine (Cys), glutathione (GSH), and glutathione peroxidase 4 (GPX4) levels were evaluated using appropriate kits. Ferroptotic markers, including Ptgs2, Chac1, SLC7A11, and SLC3A2, were analyzed by RT-qPCR, western blotting, and immunohistochemistry. ANO6 expression decreased in GIST tissues. ANO6-plasmid inhibits proliferation, induces apoptosis, and promotes pyroptosis in GIST-T1 and GIST-T1 IR cells. The ANO6-plasmid induced ferroptosis, as confirmed by enhanced lipid ROS levels, increased intracellular concentrations of total iron and Fe2+, promoted Ptgs2 and Chac1 expression, reduced Cys, GSH, and GPX4 levels, and downregulated SLC7A11 and SLC3A2 expression after in vitro and in vivo treatment with ANO6-plasmid. Moreover, the ANO6-plasmid inhibited GIST growth in vivo. Therefore, ANO6 may be a promising therapeutic target for blocking the development of GIST via the induction of apoptosis, pyroptosis, and ferroptosis.
Collapse
Affiliation(s)
- Hao Wang
- School of Public Health, Nanjing Medical University, Nanjing211166, China
- Department of Gastrointestinal Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou225001, China
| | - Wei Zhao
- Department of Gastrointestinal Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou225001, China
| | - Daorong Wang
- Department of Gastrointestinal Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou225001, China
| | - Jin Chen
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Jiangning District, Nanjing211166, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing211166, China
| |
Collapse
|
38
|
Guo S, Li Z, Liu Y, Cheng Y, Jia D. Ferroptosis: a new target for hepatic ischemia-reperfusion injury? Free Radic Res 2024; 58:396-416. [PMID: 39068663 DOI: 10.1080/10715762.2024.2386075] [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: 02/29/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024]
Abstract
Ischemia-reperfusion injury (IRI) can seriously affect graft survival and prognosis and is an unavoidable event during liver transplantation. Ferroptosis is a novel iron-dependent form of cell death characterized by iron accumulation and overwhelming lipid peroxidation; it differs morphologically, genetically, and biochemically from other well-known cell death types (autophagy, necrosis, and apoptosis). Accumulating evidence has shown that ferroptosis is involved in the pathogenesis of hepatic IRI, and targeting ferroptosis may be a promising therapeutic approach. Here, we review the pathways and phenomena involved in ferroptosis, explore the associations and implications of ferroptosis and hepatic IRI, and discuss possible strategies for modulating ferroptosis to alleviate the hepatic IRI.
Collapse
Affiliation(s)
- Shanshan Guo
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zexin Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yi Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Ying Cheng
- Department of Organ Transplantation, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Degong Jia
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
39
|
Peng Z, Ding YN, Yang ZM, Li XJ, Zhuang Z, Lu Y, Tang QS, Hang CH, Li W. Neuron-targeted liposomal coenzyme Q10 attenuates neuronal ferroptosis after subarachnoid hemorrhage by activating the ferroptosis suppressor protein 1/coenzyme Q10 system. Acta Biomater 2024; 179:325-339. [PMID: 38561074 DOI: 10.1016/j.actbio.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024]
Abstract
Subarachnoid hemorrhage (SAH) is primarily attributed to the rupture of intracranial aneurysms and is associated with a high incidence of disability and mortality. SAH disrupts the blood‒brain barrier, leading to the release of iron ions from blood within the subarachnoid space, subsequently inducing neuronal ferroptosis. A recently discovered protein, known as ferroptosis suppressor protein 1 (FSP1), exerts anti-ferroptotic effects by facilitating the conversion of oxidative coenzyme Q 10 (CoQ10) to its reduced form, which effectively scavenges reactive oxygen radicals and mitigates iron-induced ferroptosis. In our investigation, we observed an increase in FSP1 levels following SAH. However, the depletion of CoQ10 caused by SAH hindered the biological function of FSP1. Therefore, we created neuron-targeted liposomal CoQ10 by introducing the neuron-targeting peptide Tet1 onto the surface of liposomal CoQ10. Our objective was to determine whether this formulation could activate the FSP1 system and subsequently inhibit neuronal ferroptosis. Our findings revealed that neuron-targeted liposomal CoQ10 effectively localized to neurons at the lesion site after SAH. Furthermore, it facilitated the upregulation of FSP1, reduced the accumulation of malondialdehyde and reactive oxygen species, inhibited neuronal ferroptosis, and exerted neuroprotective effects both in vitro and in vivo. Our study provides evidence that supplementation with CoQ10 can effectively activate the FSP1 system. Additionally, we developed a neuron-targeted liposomal CoQ10 formulation that can be selectively delivered to neurons at the site of SAH. This innovative approach represents a promising therapeutic strategy for neuronal ferroptosis following SAH. STATEMENT OF SIGNIFICANCE: Subarachnoid hemorrhage (SAH) is primarily attributed to the rupture of intracranial aneurysms and is associated with a high incidence of disability and mortality. Ferroptosis suppressor protein 1 (FSP1), exerts anti-ferroptotic effects by facilitating the conversion of oxidative coenzyme Q 10 (CoQ10) to its reduced form, which effectively scavenges reactive oxygen radicals and mitigates iron-induced ferroptosis. In our investigation, we observed an increase in FSP1 levels following SAH. However, the depletion of CoQ10 caused by SAH hindered the biological function of FSP1. Therefore, we created neuron-targeted liposomal CoQ10. We find that it effectively localized to neurons at the lesion site after SAH and activated the FSP1/CoQ10 system. This innovative approach represents a promising therapeutic strategy for neuronal ferroptosis following SAH and other central nervous system diseases characterized by disruption of the blood-brain barrier.
Collapse
Affiliation(s)
- Zheng Peng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Yi-Nan Ding
- Department of Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
| | | | - Xiao-Jian Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Zong Zhuang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Yue Lu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Qiu-Sha Tang
- Medical School of Southeast University, Nanjing, China.
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| |
Collapse
|
40
|
Xiang W, Li L, Zhao Q, Zeng Y, Shi J, Chen Z, Gao G, Lai K. PEDF protects retinal pigment epithelium from ferroptosis and ameliorates dry AMD-like pathology in a murine model. GeroScience 2024; 46:2697-2714. [PMID: 38153666 PMCID: PMC10828283 DOI: 10.1007/s11357-023-01038-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/05/2023] [Indexed: 12/29/2023] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of irreversible vision damage among elderly individuals. There is still no efficient treatment for dry AMD. Retinal pigment epithelial (RPE) degeneration has been confirmed to play an important role in dry AMD. Recent studies have reported that ferroptosis caused by iron overload and lipid peroxidation may be the primary causes of RPE degeneration. However, the upstream regulatory molecules of RPE ferroptosis remain largely unknown. Pigment epithelium-derived factor (PEDF) is an important endogenic protective factor for the RPE. Our results showed that in the murine dry AMD model induced by sodium iodate (SI), PEDF expression was downregulated. Moreover, dry AMD-like pathology was observed in PEDF-knockout mice. Therefore, the aim of this study was to reveal the effects and mechanism of PEDF on RPE ferroptosis and investigate potential therapeutic targets for dry AMD. The results of lipid peroxidation and transmission electron microscope showed that retinal ferroptosis was significantly activated in SI-treated mice and PEDF-knockout mice. Restoration of PEDF expression ameliorated SI-induced retinal dysfunction in mice, as assessed by electroretinography and optical coherence tomography. Mechanistically, western blotting and immunofluorescence analysis demonstrated that the overexpression of PEDF could upregulate the expression of glutathione peroxidase 4 (GPX4) and ferritin heavy chain-1 (FTH1), which proved to inhibit lipid peroxidation and RPE ferroptosis induced by SI. This study revealed the novel role of PEDF in ferroptosis inhibition and indicated that PEDF might be a potential therapeutic target for dry AMD.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Longhui Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Qin Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Yongcheng Zeng
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jinhui Shi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Zitong Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Guoquan Gao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Kunbei Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China.
| |
Collapse
|
41
|
Wang R, Rao S, Zhong Z, Xiao K, Chen X, Sun X. Emerging role of ferroptosis in diabetic retinopathy: a review. J Drug Target 2024; 32:393-403. [PMID: 38385350 DOI: 10.1080/1061186x.2024.2316775] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/20/2024] [Accepted: 02/05/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Diabetic retinopathy (DR) is a significant complication of diabetes and the primary cause of blindness among working age adults globally. The development of DR is accompanied by oxidative stress, characterised by an overproduction of reactive oxygen species (ROS) and a compromised antioxidant system. Clinical interventions aimed at mitigating oxidative stress through ROS scavenging or elimination are currently available. Nevertheless, these treatments merely provide limited management over the advanced stage of the illness. Ferroptosis is a distinctive form of cell death induced by oxidative stress, which is characterised by irondependent phospholipid peroxidation. PURPOSE This review aims to synthesise recent experimental evidence to examine the involvement of ferroptosis in the pathological processes of DR, as well as to explicate the regulatory pathways governing oxidative stress and ferroptosis in retina. METHODS We systematically reviewed literature available up to 2023. RESULTS This review included 12 studies investigating the involvement of ferroptosis in DR.
Collapse
Affiliation(s)
- Ruohong Wang
- Department of Ophthalmology Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Suyun Rao
- Department of Ophthalmology Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Zheng Zhong
- Department of Ophthalmology Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Ke Xiao
- Department of Ophthalmology Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Xuhui Chen
- Department of Ophthalmology Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Xufang Sun
- Department of Ophthalmology Tongji Hospital Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| |
Collapse
|
42
|
Liu D, Liu Z, Liao H, Chen ZS, Qin B. Ferroptosis as a potential therapeutic target for age-related macular degeneration. Drug Discov Today 2024; 29:103920. [PMID: 38369100 DOI: 10.1016/j.drudis.2024.103920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/20/2024]
Abstract
Cell death plays a crucial part in the process of age-related macular degeneration (AMD), but its mechanisms remain elusive. Accumulating evidence suggests that ferroptosis, a novel form of regulatory cell death characterized by iron-dependent accumulation of lipid hydroperoxides, has a crucial role in the pathogenesis of AMD. Numerous studies have suggested that ferroptosis participates in the degradation of retinal cells and accelerates the progression of AMD. Furthermore, inhibitors of ferroptosis exhibit notable protective effects in AMD, underscoring the significance of ferroptosis as a pivotal mechanism in the death of retinal cells during the process of AMD. This review aims to summarize the molecular mechanisms of ferroptosis in AMD, enumerate potential inhibitors and discuss the challenges and future opportunities associated with targeting ferroptosis as a therapeutic strategy, providing important information references and insights for the prevention and treatment of AMD.
Collapse
Affiliation(s)
- Dongcheng Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Ziling Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Hongxia Liao
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, New York, USA.
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China; Aier Eye Hospital, Tianjin University, Tianjin, China.
| |
Collapse
|
43
|
Jiang Y, Yu Y, Pan Z, Glandorff C, Sun M. Ferroptosis: a new hunter of hepatocellular carcinoma. Cell Death Discov 2024; 10:136. [PMID: 38480712 PMCID: PMC10937674 DOI: 10.1038/s41420-024-01863-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
Ferroptosis is an iron ion-dependent, regulatory cell death modality driven by intracellular lipid peroxidation that plays a key role in the development of HCC. Studies have shown that various clinical agents (e.g., sorafenib) have ferroptosis inducer-like effects and can exert therapeutic effects by modulating different key factors in the ferroptosis pathway. This implies that targeting tumor cell ferroptosis may be a very promising strategy for tumor therapy. In this paper, we summarize the prerequisites and defense systems for the occurrence of ferroptosis and the regulatory targets of drug-mediated ferroptosis action in HCC, the differences and connections between ferroptosis and other programmed cell deaths. We aim to summarize the theoretical basis, classical inducers of ferroptosis and research progress of ferroptosis in HCC cells, clued to the treatment of HCC by regulating ferroptosis network. Further investigation of the specific mechanisms of ferroptosis and the development of hepatocellular carcinoma and interventions at different stages of hepatocellular carcinoma will help us to deepen our understanding of hepatocellular carcinoma, with a view to providing new and more precise preventive as well as therapeutic measures for patients.
Collapse
Affiliation(s)
- Yulang Jiang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yongxin Yu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ziyang Pan
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Christian Glandorff
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- University Clinic of Hamburg at the HanseMerkur Center of TCM, Hamburg, Germany
| | - Mingyu Sun
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
44
|
Liu Z, Huang J, Li D, Zhang C, Wan H, Zeng B, Tan Y, Zhong F, Liao H, Liu M, Chen ZS, Zou C, Liu D, Qin B. Targeting ZIP8 mediated ferroptosis as a novel strategy to protect against the retinal pigment epithelial degeneration. Free Radic Biol Med 2024; 214:42-53. [PMID: 38309537 DOI: 10.1016/j.freeradbiomed.2024.01.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/10/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
The degeneration of retinal pigment epithelium (RPE) plays an important role in the development of age-related macular degeneration (AMD). However, the underlying mechanism remains elusive. In this study, we identified that ZIP8, a metal-ion transporter, plays a crucial role in the degeneration of RPE cells mediated by ferroptosis. ZIP8 was found to be upregulated in patients with AMD through transcriptome analysis. Upregulated ZIP8 was also observed in both oxidative-stressed RPE cells and AMD mouse model. Importantly, knockdown of ZIP8 significantly inhibited ferroptosis in RPE cells induced by sodium iodate-induced oxidative stress. Blocking ZIP8 with specific antibodies reversed RPE degeneration and restored retinal function, improving visual loss in a mouse model of NaIO3-induced. Interestingly, the modification of the N-glycosylation sites N40, N72 and N88, but not N273, was essential for the intracellular iron accumulation mediated by ZIP8, which further led to increased lipid peroxidation and RPE death. These findings highlight the critical role of ZIP8 in RPE ferroptosis and provide a potential target for the treatment of diseases associated with retinal degeneration, including AMD.
Collapse
Affiliation(s)
- Ziling Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Institute of Biopharmaceutics and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Jianguo Huang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Deshuang Li
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Chuanhe Zhang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Huan Wan
- The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Bing Zeng
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Yao Tan
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Fuhua Zhong
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Hongxia Liao
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - MuYun Liu
- National Engineering Research Center of Foundational Technologies for CGT Industry, Shenzhen Kenuo Medical Laboratory, China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, New York, USA
| | - Chang Zou
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.
| | - Dongcheng Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China.
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China.
| |
Collapse
|
45
|
Zhang Y, Zeng Y, Huang M, Cao G, Lin L, Wang X, Cheng Q. Andrographolide attenuates sepsis-induced acute kidney injury by inhibiting ferroptosis through the Nrf2/FSP1 pathway. Free Radic Res 2024; 58:156-169. [PMID: 38478853 DOI: 10.1080/10715762.2024.2330413] [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: 09/13/2023] [Accepted: 02/04/2024] [Indexed: 03/23/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome caused by infection, which causes renal dysfunction known as sepsis-associated acute kidney injury (S-AKI). Ferroptosis is a form of lipid peroxidation dependent on iron and reactive oxygen species that differs from other forms of programmed cell death at the morphological and biochemical levels. Andrographolide (AG), a natural diterpenoid lactone compound extracted from Andrographis paniculata, has been shown to have therapeutic effects in kidney disease. In this study, we investigated the novel mechanism by which AG attenuates septic acute kidney injury by inhibiting ferroptosis in renal tubular epithelial cells (HK-2) through the Nrf2/FSP1 pathway. Cecum ligation and puncture (CLP)-induced septic rats and lipopolysaccharide (LPS)-induced HK-2 cells were used for in vivo and in vitro experiments. Firstly, in septic rats and HK-2 cells, AG effectively decreased the levels of kidney injury indicators, including blood creatinine, urea nitrogen, and markers of kidney injury such as neutrophil gelatinase-associated lipid transport protein and kidney injury molecule-1 (KIM-1). In addition, AG prevented ferroptotosis, by avoiding the accumulation of iron and lipid peroxidation, and an increase in SLC7A11 and GPX4 in AG-treated HK-2 cells. Furthermore, AG attenuated mitochondrial damage, including mitochondrial swelling, outer membrane rupture, and a reduction in mitochondrial cristae in LPS-treated HK-2 cells. Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, significantly inhibited LPS-induced ferroptosis in HK-2 cells. Importantly, our results confirm that Nrf2/FSP1 is an important pathway for ferroptosis resistance. Nrf2 siRNA hindered the effect of AG in attenuating acute kidney injury and inhibiting ferroptosis. These findings demonstrate that Nrf2/FSP1-mediated HK-2 ferroptosis is associated with AG, alleviates septic acute kidney injury, and indicates a novel avenue for therapeutic interventions in the treatment of acute kidney injury in sepsis.
Collapse
Affiliation(s)
- Yixin Zhang
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | | | - Ming Huang
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | | | - Liang Lin
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Xiaoyue Wang
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Qinghong Cheng
- The First Affiliated Hospital of Shihezi University, Shihezi, China
| |
Collapse
|
46
|
He X, Wedn A, Wang J, Gu Y, Liu H, Zhang J, Lin Z, Zhou R, Pang X, Cui Y. IUPHAR ECR review: The cGAS-STING pathway: Novel functions beyond innate immune and emerging therapeutic opportunities. Pharmacol Res 2024; 201:107063. [PMID: 38216006 DOI: 10.1016/j.phrs.2024.107063] [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: 12/07/2023] [Revised: 12/26/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Stimulator of interferon genes (STING) is a crucial innate immune sensor responsible for distinguishing pathogens and cytosolic DNA, mediating innate immune signaling pathways to defend the host. Recent studies have revealed additional regulatory functions of STING beyond its innate immune-related activities, including the regulation of cellular metabolism, DNA repair, cellular senescence, autophagy and various cell deaths. These findings highlight the broader implications of STING in cellular physiology beyond its role in innate immunity. Currently, approximately 10 STING agonists have entered the clinical stage. Unlike inhibitors, which have a maximum inhibition limit, agonists have the potential for infinite amplification. STING signaling is a complex process that requires precise regulation of STING to ensure balanced immune responses and prevent detrimental autoinflammation. Recent research on the structural mechanism of STING autoinhibition and its negative regulation by adaptor protein complex 1 (AP-1) provides valuable insights into its different effects under physiological and pathological conditions, offering a new perspective for developing immune regulatory drugs. Herein, we present a comprehensive overview of the regulatory functions and molecular mechanisms of STING beyond innate immune regulation, along with updated details of its structural mechanisms. We discuss the implications of these complex regulations in various diseases, emphasizing the importance and feasibility of targeting the immunity-dependent or immunity-independent functions of STING. Moreover, we highlight the current trend in drug development and key points for clinical research, basic research, and translational research related to STING.
Collapse
Affiliation(s)
- Xu He
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing 100191, China; Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing 100034, China
| | - Abdalla Wedn
- School of Medicine, University of Pittsburgh, 5051 Centre Avenue, Pittsburgh, PA, USA
| | - Jian Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yanlun Gu
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing 100191, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing 100191, China
| | - Hongjin Liu
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing 100034, China
| | - Juqi Zhang
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing 100191, China; Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing 100034, China
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Renpeng Zhou
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Anhui 230601, China; Department of Orthopedics and Rehabilitation, Yale University School of Medicine, New Haven CT06519, USA.
| | - Xiaocong Pang
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing 100191, China; Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing 100034, China.
| | - Yimin Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing 100191, China; Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing 100034, China.
| |
Collapse
|
47
|
Zhang Q, Xiong K. Editorial: Novel strategies to target cell death signaling in cancer and neurodegenerative diseases: new findings and mechanistic studies. Front Cell Dev Biol 2024; 12:1383301. [PMID: 38469180 PMCID: PMC10925791 DOI: 10.3389/fcell.2024.1383301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 03/13/2024] Open
Affiliation(s)
- Qi Zhang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Hunan Key Laboratory of Ophthalmology, Changsha, China
| |
Collapse
|
48
|
Upadhyay M, Bonilha VL. Regulated cell death pathways in the sodium iodate model: Insights and implications for AMD. Exp Eye Res 2024; 238:109728. [PMID: 37972750 PMCID: PMC10841589 DOI: 10.1016/j.exer.2023.109728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
The sodium iodate (NaIO3) model of increased oxidative stress recapitulates dry AMD features such as patchy RPE loss, secondary photoreceptors, and underlying choriocapillaris death, allowing longitudinal evaluation of the retinal structure. Due to the time- and dose-dependent degeneration observed in diverse animal models, this preclinical model has become one of the most studied models. The events leading to RPE cell death post- NaIO3 injection have been extensively studied, and here we have reviewed different modalities of cell death, including apoptosis, necroptosis, ferroptosis, and pyroptosis with a particular focus on findings associated with in vivo and in vitro NaIO3 studies on RPE cell death. Because the fundamental cause of vision loss in patients with dry AMD is the death of these same cells affected by NaIO3, studies using NaIO3 can provide valuable insights into RPE and photoreceptor cell death mechanisms and can help understand mechanisms behind RPE degeneration in AMD.
Collapse
Affiliation(s)
- Mala Upadhyay
- Cole Eye Institute, Ophthalmic Research, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Vera L Bonilha
- Cole Eye Institute, Ophthalmic Research, Cleveland Clinic, Cleveland, OH, 44195, USA; Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.
| |
Collapse
|
49
|
Guo J, Chen L, Ma M. Ginsenoside Rg1 Suppresses Ferroptosis of Renal Tubular Epithelial Cells in Sepsis-induced Acute Kidney Injury via the FSP1-CoQ 10- NAD(P)H Pathway. Curr Med Chem 2024; 31:2119-2132. [PMID: 37287288 DOI: 10.2174/0929867330666230607125054] [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: 12/06/2022] [Revised: 04/26/2023] [Accepted: 05/12/2023] [Indexed: 06/09/2023]
Abstract
INTRODUCTION Sepsis-induced acute kidney injury is related to an increased mortality rate by modulating ferroptosis through ginsenoside Rg1. In this study, we explored the specific mechanism of it. METHODS Human renal tubular epithelial cells (HK-2) were transfected with oe-ferroptosis suppressor protein 1 and treated with lipopolysaccharide for ferroptosis induction, and they were then treated with ginsenoside Rg1 and ferroptosis suppressor protein 1 inhibitor. Ferroptosis suppressor protein 1, CoQ10, CoQ10H2, and intracellular NADH levels in HK-2 cells were assessed by Western blot, ELISA kit, and NAD/NADH kit. NAD+/NADH ratio was also calculated, and 4-Hydroxynonal fluorescence intensity was assessed by immunofluorescence. HK-2 cell viability and death were assessed by CCK-8 and propidium iodide staining. Ferroptosis, lipid peroxidation, and reactive oxygen species accumulation were assessed by Western blot, kits, flow cytometry, and C11 BODIPY 581/591 molecular probe. Sepsis rat models were established by cecal ligation and perforation to investigate whether ginsenoside Rg1 regulated the ferroptosis suppressor protein 1-CoQ10-NAD(P)H pathway in vivo. RESULTS LPS treatment diminished ferroptosis suppressor protein 1, CoQ10, CoQ10H2, and NADH contents in HK-2 cells, while facilitating NAD+/NADH ratio and relative 4- Hydroxynonal fluorescence intensity. FSP1 overexpression inhibited lipopolysaccharideinduced lipid peroxidation in HK-2 cells via the ferroptosis suppressor protein 1-CoQ10- NAD(P)H pathway. The ferroptosis suppressor protein 1-CoQ10-NAD(P)H pathway suppressed lipopolysaccharide-induced ferroptosis in HK-2 cells. Ginsenoside Rg1 alleviated ferroptosis in HK-2 cells by regulating the ferroptosis suppressor protein 1-CoQ10- NAD(P)H pathway. Moreover, ginsenoside Rg1 regulated the ferroptosis suppressor protein 1-CoQ10-NAD(P)H pathway in vivo. CONCLUSION Ginsenoside Rg1 alleviated sepsis-induced acute kidney injury by blocking renal tubular epithelial cell ferroptosis via the ferroptosis suppressor protein 1-CoQ10- NAD(P)H pathway.
Collapse
Affiliation(s)
- Jun Guo
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, 210009, Jiangsu Province, China
- Department of Critical Care Medicine, Union Jiangbei Hospital, Huazhong University of Science and Technology, No. 111, Success Road, Caidian District, Wuhan, 430100, Hubei Province, China
| | - Long Chen
- Department of Critical Care Medicine, Union Jiangbei Hospital, Huazhong University of Science and Technology, No. 111, Success Road, Caidian District, Wuhan, 430100, Hubei Province, China
| | - Min Ma
- Department of Critical Care Medicine, Union Jiangbei Hospital, Huazhong University of Science and Technology, No. 111, Success Road, Caidian District, Wuhan, 430100, Hubei Province, China
| |
Collapse
|
50
|
Shen X, Dong P, Kong J, Sun N, Wang F, Sang L, Xu Y, Zhang M, Chen X, Guo R, Wang S, Lin Q, Jiang Z, Xu S, Zhang C, Bian Z, Wang W, Guo R. Targeted single-cell RNA sequencing analysis reveals metabolic reprogramming and the ferroptosis-resistant state in hematologic malignancies. Cell Biochem Funct 2023; 41:1343-1356. [PMID: 37823726 DOI: 10.1002/cbf.3869] [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: 07/11/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023]
Abstract
Hematologic malignancies are the most common hematopoietic diseases and a major public health concern. However, the mechanisms underlying myeloid tumors remain unknown owing to the intricate interplay between mutations and diverse clonal evolution patterns, as evidenced by the analysis of bulk cell-derived omics data. Several single-cell omics techniques have been used to characterize the hierarchies and altered immune microenvironments of hematologic malignancies. The comprehensive single-cell atlas of hematologic malignancies provides novel opportunities for personalized combinatorial targeted treatments, avoiding unwanted chemo-toxicity. In the present study, we performed transcriptome sequencing by combining single-cell RNA sequencing (scRNA-seq) with a targeted oncogenic gene panel for acute myeloid leukemia, overcoming the limitations of scRNA-seq in detecting oncogenic mutations. The distribution of oncogenic IDH1, IDH2, and KRAS mutations in each cell type was identified in the bone marrow (BM) samples of each patient. Our findings suggest that ferroptosis and metabolic reprogramming are involved in the tumorigenesis and chemotherapy resistance of oncogenic mutation-carrying cells. Biological progression via IDH1, IDH2, and KRAS mutations arrests hematopoietic maturation. Our study findings provide a rationale for using primary BM cells for personalized treatment in clinical settings.
Collapse
Affiliation(s)
- Xiaohui Shen
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peiyuan Dong
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Kong
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nannan Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lina Sang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Xu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengmeng Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoli Chen
- Jiangxi Health Commission Key Laboratory of Leukemia, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, China
| | - Rong Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuya Wang
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Quande Lin
- Department of Hematology, The Afliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shan Xu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Congli Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhilei Bian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Weimin Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|