51
|
Song Y, Hai E, Zhang N, Zhang Y, Wang J, Han X, Zhang J. Oocyte transcriptomes and follicular fluid proteomics of ovine atretic follicles reveal the underlying mechanisms of oocyte degeneration. BMC Genomics 2025; 26:97. [PMID: 39893388 PMCID: PMC11786490 DOI: 10.1186/s12864-025-11291-9] [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: 05/27/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND In mammals, female fertility is influenced by the result of follicular development (ovulation or atresia). Follicular atresia is a complex physiological process that results in the degeneration of oocytes from the ovary. However, the molecular mechanisms of oocyte degeneration and key protein markers of follicular atresia remain unclear. In this study, we investigated the complex transcriptional regulatory mechanisms and protein profiles in oocytes and follicular fluid in atretic follicle stages using single-cell RNA sequencing and tandem mass tag proteomics. RESULTS First, through paired analysis of different follicle development stages, we identified 175 atresia-specific genes and eight candidate oocyte-secreted factors, including PKG1, YTHDF2, and MYC. Meanwhile, we also characterized unique features of the oocyte transcriptional landscape in the atretic follicle stage that displayed cell death-related transcriptional changes and mechanisms, such as autophagy (TBK1 and IRS4), necroptosis (PKR), and apoptosis (MARCKS). Moreover, we identified atresia-specific genes, namely FTH1, TF, and ACSL4, which may participate in regulation of oocyte ferroptosis in atretic follicles through a series of mechanisms including ferritinophagy, ferritin transport, and lipid metabolism. Additionally, we uncovered 333 differentially expressed proteins that may coordinate follicular atresia and revealed key pathways, such as negative regulation of angiogenesis, metabolic pathways, and transcription and mRNA splicing, that lead to oocyte degeneration. Finally, by combining transcriptome and proteomics analyses, we identified two oocyte-secreted biomarkers, PGK1 and ANGPT2, that may be associated with follicular atresia. CONCLUSIONS In conclusion, our work offers a thorough characterization of oocyte transcription mechanism and follicular fluid protein changes in ovine atretic follicles, which offers a crucial reference for analyzing the mechanism of follicular atresia and establishing an oocyte quality assessment system in sheep.
Collapse
Affiliation(s)
- Yukun Song
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Erhan Hai
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Nan Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Yu Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Junlan Wang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Xitong Han
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Jiaxin Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China.
| |
Collapse
|
52
|
Chen Y, Liu X, Chen B, Zhu F, Wang Z, Cheong KL, Ye S, Zhong S, Chen J. Selenium nanoparticles decorated by fucoidan induce ferroptosis in HepG2 cells. Int J Biol Macromol 2025; 289:138841. [PMID: 39701247 DOI: 10.1016/j.ijbiomac.2024.138841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
Unlike apoptosis, necrosis and autophagy, ferroptosis is a novel type of regulated cell death, and the mechanism by which selenium nanoparticles induce ferroptosis in cancer cells has rarely been investigated. To investigate the mechanism of inhibition of HepG2 cell proliferation by fucoidan-selenium nanoparticles (FD-SeNPs) by inducing ferroptosis. The mechanism was explored by detecting ROS, MDA, GSH and Fe2+ and utilizing TEM and Western blot assay. The results showed that FD-SeNPs increased intracellular ROS, MDA and Fe2+ levels and decreased GSH levels. Moreover, HepG2 cells treated with FD-SeNPs showed mitochondrial shrinkage, volume reduction and mitochondrial cristae breakage. The ability to reverse the changes in the above indexes after Ferrostatin-1 (Fer-1) intervention suggests that FD-SeNPs inhibit HepG2 cell proliferation by inducing cells to undergo ferroptosis. Further mechanistic studies revealed that FD-SeNPs decreased the expression of Nrf2, HO-1, SLC7A11 (xCT), GCLC and GPX4 proteins to promote lipid peroxidation in HepG2 cells. Moreover, FD-SeNPs could disrupt intracellular iron homeostasis by up-regulating transferrin protein and down-regulating SLC40A1 and Ferritin proteins, suggesting that FD-SeNPs induced cells to undergo ferroptosis by regulating proteins related to lipid peroxidation and iron homeostasis. This study provides theoretical data for reference in applying FD-SeNPs in developing anti-cancer clinical drugs.
Collapse
Affiliation(s)
- Yanzhe Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Xiaofei Liu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Bowen Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Feifei Zhu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Zhuo Wang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Kit-Leong Cheong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Shengquan Ye
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Saiyi Zhong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Jianping Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China; Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan 528000, China.
| |
Collapse
|
53
|
Tu Y, Tang E, Ye H, Xiang Q, Ye Z, Hao Y, Liao W. Flammulina Velutipes polysaccharides ameliorate cisplatin-induced acute kidney injury in mice via regulation of gut microbiota and Ferroptosis pathway. Int J Biol Macromol 2025; 290:138526. [PMID: 39706410 DOI: 10.1016/j.ijbiomac.2024.138526] [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: 12/22/2023] [Revised: 11/17/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024]
Abstract
Acute kidney injury (AKI) is a common and serious clinical complication with high incidence. Polysaccharides extracted from Flammulina velutipes (FVPs) have been proven to possess anti-inflammatory and antioxidant properties. The present study aimed to investigate the ameliorative effect and mechanism of FVPs on cisplatin (CDPP)-induced AKI. The results of our study revealed that FVPs improved CDPP-induced AKI in mice as indicated by decreasing serum creatinine and urea levels and down-regulating the mRNA expression of IL-6 and TNF-α. Moreover, FVPs modified the composition of gut microorganisms and increased the content of short-chain fatty acids (SCFAs). Additionally, kidney metabolomics analysis demonstrated enrichment of the ferroptosis metabolic pathway. Furthermore, FVPs suppressed ferroptosis as shown by increasing levels of GSH, GPX4, and SLC7A11, while reducing the arachidonic acid level. In conclusion, FVPs were confirmed to ameliorate CDPP-induced AKI in the present study. FVPs can modify the composition of the gut microbiota to promote the production of SCFAs, as well as modulate renal metabolism and inhibit ferroptosis.
Collapse
Affiliation(s)
- Yali Tu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China; Leshan Vocational and Technical College, Leshan, China
| | - Enhui Tang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Huarui Ye
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qianru Xiang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zichong Ye
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuting Hao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China.
| |
Collapse
|
54
|
Lai L, Tan M, Hu M, Yue X, Tao L, Zhai Y, Li Y. Important molecular mechanisms in ferroptosis. Mol Cell Biochem 2025; 480:639-658. [PMID: 38668809 DOI: 10.1007/s11010-024-05009-w] [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: 12/26/2023] [Accepted: 04/07/2024] [Indexed: 02/19/2025]
Abstract
Ferroptosis is a type of cell death that is caused by the oxidation of lipids and is dependent on the presence of iron. It was first characterized by Brent R. Stockwell in 2012, and since then, research in the field of ferroptosis has rapidly expanded. The process of ferroptosis-induced cell death is genetically, biochemically, and morphologically distinct from other forms of cellular death, such as apoptosis, necroptosis, and non-programmed cell death. Extensive research has been devoted to comprehending the intricate process of ferroptosis and the various factors that contribute to it. While the majority of these studies have focused on examining the effects of lipid metabolism and mitochondria on ferroptosis, recent findings have highlighted the significant involvement of signaling pathways and associated proteins, including Nrf2, P53, and YAP/TAZ, in this process. This review provides a concise summary of the crucial signaling pathways associated with ferroptosis based on relevant studies. It also elaborates on the drugs that have been employed in recent years to treat ferroptosis-related diseases by targeting the relevant signaling pathways. The established and potential therapeutic targets for ferroptosis-related diseases, such as cancer and ischemic heart disease, are systematically addressed.
Collapse
Affiliation(s)
- Lunmeng Lai
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Menglei Tan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Mingming Hu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Xiyue Yue
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Lulu Tao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Yanru Zhai
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Yunsen Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China.
| |
Collapse
|
55
|
Matboli M, Hamady S, Saad M, Khaled R, Khaled A, Barakat EMF, Sayed SA, Agwa S, Youssef I. Innovative approaches to metabolic dysfunction-associated steatohepatitis diagnosis and stratification. Noncoding RNA Res 2025; 10:206-222. [PMID: 40248839 PMCID: PMC12004009 DOI: 10.1016/j.ncrna.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/08/2024] [Accepted: 10/10/2024] [Indexed: 01/03/2025] Open
Abstract
The global rise in Metabolic dysfunction-associated steatotic liver disease (MASLD)/Metabolic dysfunction-associated steatohepatitis (MASH) highlights the urgent necessity for noninvasive biomarkers to detect these conditions early. To address this, we endeavored to construct a diagnostic model for MASLD/MASH using a combination of bioinformatics, molecular/biochemical data, and machine learning techniques. Initially, bioinformatics analysis was employed to identify RNA molecules associated with MASLD/MASH pathogenesis and enriched in ferroptosis and exophagy. This analysis unveiled specific networks related to ferroptosis (GPX4, LPCAT3, ACSL4, miR-4266, and LINC00442) and exophagy (TSG101, HGS, SNF8, miR-4498, miR-5189-5p, and CTBP1-AS2). Subsequently, serum samples from 400 participants (151 healthy, 150 MASH, and 99 MASLD) underwent biochemical and molecular analysis, revealing significant dyslipidemia, impaired liver function, and disrupted glycemic indicators in MASLD/MASH patients compared to healthy controls. Molecular analysis indicated increased expression of LPCAT3, ACSL4, TSG101, HGS, and SNF8, alongside decreased GPX4 levels in MASH and MASLD patients compared to controls. The expression of epigenetic regulators from both networks (miR-4498, miR-5189-5p, miR-4266, LINC00442, and CTBP1-AS2) significantly differed among the studied groups. Finally, supervised machine learning models, including Neural Networks and Random Forest, were applied to molecular signatures and clinical/biochemical data. The Random Forest model exhibited superior performance, and molecular features effectively distinguished between the three studied groups. Clinical features, particularly BMI, consistently served as discriminatory factors, while biochemical features exhibited varying discriminant behavior across MASH, MASLD, and control groups. Our study underscores the significant potential of integrating diverse data types to enable early detection of MASLD/MASH, offering a promising approach for non-invasive diagnostic strategies.
Collapse
Affiliation(s)
- Marwa Matboli
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt
- Faculty of Oral & Dental Medicine, Misr International University, Qalyubiyya Governorate, Egypt
| | - Shaimaa Hamady
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Maha Saad
- Basic Sciences Department, Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| | - Radwa Khaled
- Basic Sciences Department, Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University & Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| | - Abdelrahman Khaled
- Bioinformatics Group, Center of Informatics Sciences (CIS), School of Information Technology and Computer Sciences, Nile University, Giza, Egypt
| | - Eman MF. Barakat
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sayed Ahmed Sayed
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - SaraH.A. Agwa
- Clinical Pathology and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo, 11382, Egypt
| | - Ibrahim Youssef
- Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Egypt
| |
Collapse
|
56
|
He N, Yuan D, Luo M, Xu Q, Wen Z, Wang Z, Zhao J, Liu Y. Ferroptosis contributes to immunosuppression. Front Med 2025; 19:1-22. [PMID: 39560919 DOI: 10.1007/s11684-024-1080-8] [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: 12/26/2023] [Accepted: 04/18/2024] [Indexed: 11/20/2024]
Abstract
As a novel form of cell death, ferroptosis is mainly regulated by the accumulation of soluble iron ions in the cytoplasm and the production of lipid peroxides and is closely associated with several diseases, including acute kidney injury, ischemic reperfusion injury, neurodegenerative diseases, and cancer. The term "immunosuppression" refers to various factors that can directly harm immune cells' structure and function and affect the synthesis, release, and biological activity of immune molecules, leading to the insufficient response of the immune system to antigen production, failure to successfully resist the invasion of foreign pathogens, and even organ damage and metabolic disorders. An immunosuppressive phase commonly occurs in the progression of many ferroptosis-related diseases, and ferroptosis can directly inhibit immune cell function. However, the relationship between ferroptosis and immunosuppression has not yet been published due to their complicated interactions in various diseases. Therefore, this review deeply discusses the contribution of ferroptosis to immunosuppression in specific cases. In addition to offering new therapeutic targets for ferroptosis-related diseases, the findings will help clarify the issues on how ferroptosis contributes to immunosuppression.
Collapse
Affiliation(s)
- Nina He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Dun Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Minjie Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Qing Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Zhongchi Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Ziqin Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China.
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China.
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China.
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, China.
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, China.
- National Medicine Functional Experimental Teaching Center, Changsha, 410008, China.
| |
Collapse
|
57
|
Liang S, Wu Y, Zhang R, Xu L, Xie F. TNFSF9 Silence Impedes Cerebral Ischemia-Reperfusion Injury via Modulating SLC3A2 Expression in Brain Microvascular Endothelial Cells. J Mol Neurosci 2025; 75:12. [PMID: 39856410 DOI: 10.1007/s12031-025-02310-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI), which stays unresolved in the clinic, occurs after recanalization of blood vessels serving brain tissues in acute ischemic stroke patients and can result in massive brain cell death, and cell ferroptosis contributes greatly to this process. Our research firstly found that TNFSF9 expression harbored diagnostic value on CIRI patients and intended to further investigate its regulatory mechanism in CIRI, which might facilitate its diagnostic and therapeutic application in the clinic. The level of TNSF9 mRNA was augmented in the plasma of CIR patients, and its silence impeded ferroptosis, apoptosis, and release of inflammatory mediators of BMECs with OGD/R treatment. Besides, SP1 positively regulated TNFSF9 expression as one of its transcription factors, and TNFSF9 overexpression reversed SP1 silence-mediated inhibition on ferroptosis, apoptosis, and release of inflammatory mediators in OGD/R-treated BMECs. In addition, silencing SLC3A2 could neutralize the benefit effects of TNFSF9 downregulation on BMECs under OGD/R context in vitro, and silencing TNFSF9 neutralized necrotic volumes in rat brain induced by CIRI via modulating SLC3A2 expression in vivo. TNFSF9 regulated by SP1 aggravated CIRI via boosting ferroptosis, apoptosis, and release of inflammatory mediators of BMECs under OGD/R situation by suppressing SLC3A2 expression in vitro and in vivo.
Collapse
Affiliation(s)
- Shunli Liang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310005, Zhejiang, China
- The Second Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou City, 310053, Zhejiang, China
| | - You Wu
- Department of Neurology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310005, Zhejiang, China
| | - Rongbo Zhang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310005, Zhejiang, China
| | - Linsheng Xu
- Department of Neurology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310005, Zhejiang, China
| | - Fangping Xie
- Department of Special Examination, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, No. 305 Tianmushan Road, Hangzhou City, 310013, Zhejiang, China.
| |
Collapse
|
58
|
Pedrera L, Prieto Clemente L, Dahlhaus A, Lotfipour Nasudivar S, Tishina S, Olmo González D, Stroh J, Yapici FI, Singh RP, Grotehans N, Langer T, García-Sáez AJ, von Karstedt S. Ferroptosis triggers mitochondrial fragmentation via Drp1 activation. Cell Death Dis 2025; 16:40. [PMID: 39863602 PMCID: PMC11762985 DOI: 10.1038/s41419-024-07312-2] [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: 07/09/2024] [Revised: 11/21/2024] [Accepted: 12/16/2024] [Indexed: 01/27/2025]
Abstract
Constitutive mitochondrial dynamics ensure quality control and metabolic fitness of cells, and their dysregulation has been implicated in various human diseases. The large GTPase Dynamin-related protein 1 (Drp1) is intimately involved in mediating constitutive mitochondrial fission and has been implicated in mitochondrial cell death pathways. During ferroptosis, a recently identified type of regulated necrosis driven by excessive lipid peroxidation, mitochondrial fragmentation has been observed. Yet, how this is regulated and whether it is involved in ferroptotic cell death has remained unexplored. Here, we provide evidence that Drp1 is activated upon experimental induction of ferroptosis and promotes cell death execution and mitochondrial fragmentation. Using time-lapse microscopy, we found that ferroptosis induced mitochondrial fragmentation and loss of mitochondrial membrane potential, but not mitochondrial outer membrane permeabilization. Importantly, Drp1 accelerated ferroptotic cell death kinetics. Notably, this function was mediated by the regulation of mitochondrial dynamics, as overexpression of Mitofusin 2 phenocopied the effect of Drp1 deficiency in delaying ferroptosis cell death kinetics. Mechanistically, we found that Drp1 is phosphorylated and activated after induction of ferroptosis and that it translocates to mitochondria. Further activation at mitochondria through the phosphatase PGAM5 promoted ferroptotic cell death. Remarkably, Drp1 depletion delayed mitochondrial and plasma membrane lipid peroxidation. These data provide evidence for a functional role of Drp1 activation and mitochondrial fragmentation in the acceleration of ferroptotic cell death, with important implications for targeting mitochondrial dynamics in diseases associated with ferroptosis.
Collapse
Affiliation(s)
- Lohans Pedrera
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Laura Prieto Clemente
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alina Dahlhaus
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sara Lotfipour Nasudivar
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Sofya Tishina
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Daniel Olmo González
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- University of Barcelona, Barcelona, Spain
| | - Jenny Stroh
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Fatma Isil Yapici
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Randhwaj Pratap Singh
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Nils Grotehans
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Thomas Langer
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ana J García-Sáez
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany.
- Institute for Genetics, University of Cologne, Cologne, Germany.
- Max Planck Institute of Biophysics, Frankfurt, Germany.
| | - Silvia von Karstedt
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany.
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
59
|
Alatawi AD, Venkatesan K, Asseri K, Paulsamy P, Alqifari SF, Ahmed R, Nagoor Thangam MM, Sirag N, Qureshi AA, Elsayes HA, Faried Bahgat Z, Bahnsawy NSM, Prabahar K, Dawood BMAE. Targeting Ferroptosis in Rare Neurological Disorders Including Pediatric Conditions: Innovations and Therapeutic Challenges. Biomedicines 2025; 13:265. [PMID: 40002678 PMCID: PMC11853599 DOI: 10.3390/biomedicines13020265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/09/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Ferroptosis, characterized by iron dependency and lipid peroxidation, has emerged as a key mechanism underlying neurodegeneration in rare neurological disorders. These conditions, often marked by significant therapeutic gaps and high unmet medical needs, present unique challenges for intervention development. This review examines the involvement of ferroptosis in rare neurological disease pathogenesis, focusing on its role in oxidative damage and neuronal dysfunction. We explore recent pharmacological advancements, including iron chelators, lipid peroxidation blockers, and antioxidant-based strategies, designed to target ferroptosis. While these approaches show promise, challenges such as disease heterogeneity, limited diagnostic tools, and small patient cohorts hinder progress. Furthermore, we discuss the translational and regulatory barriers to implementing ferroptosis-based therapies in clinical practice. By addressing these obstacles and fostering innovative solutions, this review underscores the potential of ferroptosis-targeting strategies to revolutionize treatment paradigms for rare neurological disorders.
Collapse
Affiliation(s)
- Ahmed D. Alatawi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Krishnaraju Venkatesan
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62521, Saudi Arabia; (K.A.); (A.A.Q.)
| | - Khalid Asseri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62521, Saudi Arabia; (K.A.); (A.A.Q.)
| | - Premalatha Paulsamy
- College of Nursing, Mahalah Branch for Girls, King Khalid University, Abha 62521, Saudi Arabia;
| | - Saleh F. Alqifari
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.F.A.); (K.P.)
| | - Rehab Ahmed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (N.S.)
| | | | - Nizar Sirag
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (N.S.)
| | - Absar A. Qureshi
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62521, Saudi Arabia; (K.A.); (A.A.Q.)
| | - Hala Ahmed Elsayes
- Department of Psychiatric and Mental Health Nursing, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Psychiatric and Mental Health, Faculty of Nursing, Tanta University, Tanta 31527, Egypt
| | - Zeinab Faried Bahgat
- Department of Medical-Surgical Nursing, Faculty of Nursing, Tanta University, Tanta 31527, Egypt;
- Department of Medical-Surgical Nursing, College of Nursing, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center, Al-Ahsa 31982, Saudi Arabia
| | - Nesren S. M. Bahnsawy
- Department of Pediatric Nursing, College of Nursing, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh 11481, Saudi Arabia;
- Department of Pediatric Nursing, Faculty of Nursing, Cairo University, Giza 12613, Egypt
| | - Kousalya Prabahar
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.F.A.); (K.P.)
| | - Basma Mahmoud Abd Elhamid Dawood
- Department of Pediatric Nursing, Faculty of Nursing, Tanta University, Tanta 31527, Egypt;
- Department of Pediatric Nursing, College of Nursing, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center, Al-Ahsa 31982, Saudi Arabia
| |
Collapse
|
60
|
Yu J, Yang H, Wang J, Huang Z, Chen S, Zhao H, Wang J, Wang Z. Comprehensive analysis of histophysiology, transcriptomics and metabolomics in goslings exposed to gossypol acetate: unraveling hepatotoxic mechanisms. Front Vet Sci 2025; 12:1527284. [PMID: 39906302 PMCID: PMC11792171 DOI: 10.3389/fvets.2025.1527284] [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: 11/13/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025] Open
Abstract
Cottonseed meal is a promising alternative to soybean meal in poultry feed, but concerns over free gossypol limit its use. Although the general toxicity of free gossypol is well-known, its specific effects on the liver-the primary site where it accumulates-are less thoroughly studied, particularly at the molecular level. This study investigated the hepatotoxic effects of gossypol acetate (GA) on goslings through a comprehensive analysis combining morphology, transcriptomics, and metabolomics. Forty-eight 7-day-old male goslings with similar body weight (BW) were randomly assigned to two groups: a control group, receiving a saline solution (0.9%, 2.5 mL/kg BW), and a GA-treated group, administered GA at 50 mg/kg BW orally for 14 days. Histological analysis revealed signs of liver damage, including granular degeneration, hepatocyte enlargement, necrosis, and mitochondrial injury. Transcriptomic analysis identified 1,137 differentially expressed genes, with 702 upregulated and 435 downregulated. Key affected pathways included carbon metabolism, glycolysis/gluconeogenesis, pyruvate metabolism, propanoate metabolism, TCA cycle, fatty acid degradation, primary bile acid biosynthesis, tryptophan metabolism, cysteine and methionine metabolism, focal adhesion, and the PPAR signaling pathway. Metabolomic analysis revealed 109 differential metabolites, 82 upregulated and 27 downregulated, implicating disruptions in linoleic acid metabolism, arachidonic acid metabolism, cAMP signaling, and serotonergic synapse pathways. Overall, GA-induced hepatotoxicity involves impaired energy production, disrupted lipid metabolism, and abnormal liver focal adhesion, leading to liver cell dysfunction. These findings highlight the vulnerability of mitochondria and critical metabolic pathways, providing insights into the molecular mechanisms of GA toxicity and guiding future studies on mitigating GA-induced liver damage in goslings.
Collapse
Affiliation(s)
- Jun Yu
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Haiming Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jian Wang
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
| | - Zixin Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Hongchang Zhao
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
| | - Jun Wang
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
| | - Zhiyue Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
61
|
Yuan Q, Wang M, Zhang Z, Wang R, Wang D, Sang Z, Zhao P, Liu X, Zhu X, Liang G, Fan H, Wang D. The ameliorative effects of melatonin against BDE-47-induced hippocampal neuronal ferroptosis and cognitive dysfunction through Nrf2-Chaperone-mediated autophagy of ACSL4 degradation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117542. [PMID: 39700775 DOI: 10.1016/j.ecoenv.2024.117542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Recent studies demonstrate that lipid peroxidation-induced ferroptosis participates in 2,2',4,4'-tetrabromodiphenyl ether (BDE-47)-evoked neurotoxicity and cognitive dysfunction. Melatonin has been indicated to confer neuroprotection against brain diseases via its potent anti-ferroptotic effects. Therefore, this study aims to explore whether melatonin can mitigate BDE-47-elicited cognitive impairment via suppressing ferroptosis, and further delineate the underlying mechanisms. Our results found that melatonin administration effectively inhibited BDE-47-induced ferroptosis in mice hippocampi and murine hippocampal neuronal HT-22 cells. Acyl-CoA synthetase long-chain family member 4 (ACSL4), a key lipid metabolism enzyme dictating ferroptosis sensitivity, accompanied by higher MDA and lipid reactive oxygen species (ROS), was remarkably increased under BDE-47 stress, while melatonin supplementation could suppress the elevated ACSL4 in vivo and in vitro. Furthermore, melatonin facilitated lysosomal ACSL4 degradation through enhancing lysosome-associated membrane protein type 2a (LAMP2a) expression and chaperone-mediated autophagy (CMA) activity, while LAMP2a knockdown abrogated the positive effects of melatonin on ACSL4 elimination in BDE-47-treated HT-22 cells. Moreover, nuclear factor erythroid 2-related factor 2 (Nrf2) activation by melatonin contributed to LAMP2a upregulation and CMA of ACSL4 and subsequent neuronal ferroptosis. Importantly, melatonin, CMA activator CA77.1, and ACSL4 inhibitor rosiglitazone (RSG) administration substantially attenuated neuronal/synaptic injury and cognitive deficits following BDE-47 exposure. Taken together, these findings revealed that melatonin could prevent BDE-47-provoked ferroptosis in the hippocampal neurons and mitigate cognitive dysfunction by facilitating ACSL4 degradation via Nrf2-chaperone-mediated autophagy. Therefore, melatonin might be a potential candidate for treating BDE-47-elicited neurotoxicity and neurobehavioral disorder.
Collapse
Affiliation(s)
- Quan Yuan
- Henan Province Rongkang Hospital, Luoyang, China
| | - Mingwei Wang
- The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Zhaoxiang Zhang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Ruofei Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Dechao Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Zichun Sang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Pu Zhao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoli Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoying Zhu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Gaofeng Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China.
| | - Dongmei Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
62
|
Zhang Y, Huang R, Liu X, Cai M, Su M, Cheng Y, Jiang J, Wang X, Peng D. Taohong siwu decoction ameliorates abnormal uterine bleeding via inhibiting ACSL4-mediated ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119130. [PMID: 39566864 DOI: 10.1016/j.jep.2024.119130] [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: 08/30/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Taohong Siwu Decoction (TSD) is a well-known traditional Chinese medicine (TCM) prescription. It consists of six crude herbs, including Rehmannia glutinosa Libosch, Paeonia lactiflora Pall, Angelica sinensis (Oliv.) Diels, Ligusticum chuanxiong Hort., Prunus persica (L.) Batsch, Cauthamus tinctorius L. It has been used to treat blood stasis syndrome in Chinese clinics for thousands of years. According to recent research, TSD may be useful in the management of abnormal uterine bleeding (AUB). The aim of the present study is to investigate the possible mechanism of TSD on AUB after drug-induced incomplete abortion. AIM OF THE STUDY To investigate whether TSD could be effective in ameliorating AUB through inhibiting acyl-CoA synthetase long-chain family member 4 (ACSL4)-mediated ferroptosis. MATERIALS AND METHODS An incomplete medical aborting model was established and Ishikawa cell lines were utilized in vitro. The quantity of uterine bleeding was measured by alkaline hemoglobin photometry. Pathological results were observed by hematoxylin-eosin staining (HE). Mitochondrial morphology and function were measured by transmission electron microscopy. The related protein and mRNA were detected by western blot, the real-time reverse transcriptase-polymerase chain reaction (RT-qPCR). We used knockdown and overexpression of ACSL4 to investigate the influence of ferroptosis in Ishikawa cells and the impact of TSD on ferroptosis. RESULTS TSD dramatically reduced the amount and duration of bleeding as well as the endometrial inflammation of AUB. TSD improved mitochondrial characteristics, decreased ACSL4 protein and mRNA levels. The ferroptosis marker glutathione (GSH) levels were increased, on the contrary, reactive oxygen species (ROS) and iron levels decreased when TSD intervened. TSD decreased levels of the inflammatory factors and the oxidative products. CONCLUSION TSD alleviated endometrial inflammation by inhibiting ACSL4-mediated ferroptosis and exerts a protective effect of AUB.
Collapse
Affiliation(s)
- Yanyan Zhang
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China
| | - Rong Huang
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Xiaochuang Liu
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China.
| | - Ming Cai
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230001, PR China
| | - Mengyu Su
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Yao Cheng
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Juanjuan Jiang
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Xuekai Wang
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Daiyin Peng
- Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| |
Collapse
|
63
|
Chen W, Yang W, Meng B, Wang X, Duan H, Xu Q, Li H. Validation and the role of PDK4 relevant to ferroptosis in degenerative lumbar disc disease. J Orthop Surg Res 2025; 20:30. [PMID: 39794775 PMCID: PMC11724506 DOI: 10.1186/s13018-024-05293-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/18/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Ferroptosis was involved in the pathogenesis of intervertebral disc degeneration (IVDD). However, the exact mechanism of IVDD associated with ferroptosis still required deeper studies. METHOD The differentially expressed genes (DEGs) in rat lumbar disc tissue between the control and IVDD group treated with IL-1β were detected by RNA sequencing (RNA-seq). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed on DEGs. We further screened the differential expressed ferroptosis-related genes (DEFRGs). Besides, a protein-protein interaction (PPI) network of DEFRGs was constructed by STRING database. The Cytoscape database identified significant modules and the hub genes. The loss function of PDK4 by siRNA inference was investigated in NPCs by CCK8 assay, ELISA assay, and the analysis of ferroptosis indicators. RESULT DEGs were identified using RNA-seq. KEGG pathway analysis showed that these genes were mainly involved in Parkinson's disease, oxytocin signaling pathway, calcium ion signaling pathway, AMPK signaling pathway, and glucagon signaling pathway. Eight hub genes (including LDHA, PKM, EP300, EGFR, EGLN1, SCD, PDK4, and FABP4) were found by the PPI network and Cytoscape on a total of 25 ferroptosis-related genes that were identified in rat lumbar disc tissue after IVDD treatment. PDK4 silencing promoted NPCS proliferation, decreased the levels of the proinflammatory factors, and suppressed ferroptosis. CONCLUSION The study suggested the potential roles of ferroptosis-related genes in IVDD and further revealed the role of PDK4 in the progression of IVDD.
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Wanliang Yang
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Bin Meng
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Xingkun Wang
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Heng Duan
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Qian Xu
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
- Cheeloo College of Medicine, Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China
| | - Hao Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
64
|
Li Q, Yin J, Lin Q, He J, Shi X, Nie H. Integrated Analysis and Validation of Ferroptosis-Related Genes Associated with Ischemia/Reperfusion Injury in Lung Transplantation. J Inflamm Res 2025; 18:251-270. [PMID: 39802518 PMCID: PMC11724631 DOI: 10.2147/jir.s489827] [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: 09/02/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025] Open
Abstract
Background Lung transplantation is the only effective therapeutic option for patients with end-stage lung disease. However, ischemia/reperfusion injury (IRI) during transplantation is a leading cause of primary graft dysfunction (PGD). Ferroptosis, a form of iron-dependent cell death driven by lipid peroxidation, has been implicated in IRI across various organs. This study aims to explore the role of ferroptosis in lung transplantation-related ischemia/reperfusion injury and to identify its potential molecular mechanisms through bioinformatics analysis. Methods Transcriptome data from lung transplant patients were obtained from the Gene Expression Omnibus (GEO) database. Ferroptosis-related differentially expressed genes (FRGs) were identified by analyzing gene expression profiles before and after reperfusion. Weighted gene co-expression network analysis (WGCNA) was used to identify module genes, and overlapping genes were further analyzed using two machine learning algorithms. The CIBERSORT algorithm was applied to assess immune cell infiltration, while Mendelian randomization (MR) analysis was used to investigate causal relationships between candidate genes and PGD. Finally, Consensus clustering based on FRGs was performed to identify subtypes. Results We identified four candidate genes associated with ferroptosis during lung reperfusion: tumor necrosis factor alpha-induced protein 3 (TNFAIP3), C-X-C motif chemokine ligand 2 (CXCL2), neural precursor cell expressed developmentally down-regulated 4-like (NEDD4L), and sestrin 2 (SESN2). These genes were closely associated with immune cell infiltration. MR analysis suggested that SESN2 might play a protective role against PGD. Additionally, consensus clustering revealed distinct immune infiltration patterns across subtypes, providing insights for personalized therapeutic approaches to lung ischemia/reperfusion injury (LIRI). Conclusion This study highlights TNFAIP3, CXCL2, NEDD4L, and SESN2 as candidate genes associated with ferroptosis during LIRI, with SESN2 potentially protecting against PGD. These findings offer promising therapeutic targets for preventing LIRI and improving outcomes in lung transplantation.
Collapse
Affiliation(s)
- Qingqing Li
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People’s Republic of China
| | - Jing Yin
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People’s Republic of China
| | - Qibin Lin
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People’s Republic of China
| | - Jilong He
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People’s Republic of China
| | - Xiu Shi
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People’s Republic of China
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People’s Republic of China
| |
Collapse
|
65
|
Liu Y, Li T, Niu C, Yuan Z, Sun S, Liu D. Hyperoxia-activated Nrf2 regulates ferroptosis in intestinal epithelial cells and intervenes in inflammatory reaction through COX-2/PGE2/EP2 pathway. Mol Med 2025; 31:1. [PMID: 39754066 PMCID: PMC11697811 DOI: 10.1186/s10020-024-00993-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/07/2024] [Indexed: 01/07/2025] Open
Abstract
The lack of knowledge about the mechanism of hyperoxia-induced intestinal injury has attracted considerable attention, due to the potential for this condition to cause neonatal complications. This study aimed to explore the relationship between hyperoxia-induced oxidative damage and ferroptosis in intestinal tissue and investigate the mechanism by which hyperoxia regulates inflammation through ferroptosis. The study systematically evaluated the effects of hyperoxia on oxidative stress, mitochondrial damage, ferroptosis, and inflammation of intestinal epithelial cells both in vitro and in vivo. The results showed that ferroptosis was involved in intestinal oxidative damage caused by hyperoxia and was regulated by Nrf2. Moreover, hyperoxia-induced oxidative damage regulated inflammation through ferroptosis by upregulating the COX-2/PGE2/EP2 signaling pathway. These findings have important implications for future clinical prevention and therapeutic approaches to neonatal organ injury caused by hyperoxia treatment.
Collapse
Affiliation(s)
- Yanping Liu
- Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, ShengJing Hospital of China Medical University, SanHao Street No. 36, HePing District, Shenyang, 110000, Liaoning, China
| | - Tianming Li
- Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, ShengJing Hospital of China Medical University, SanHao Street No. 36, HePing District, Shenyang, 110000, Liaoning, China
- Department of Pathology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, People's Republic of China
| | - Changping Niu
- Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, ShengJing Hospital of China Medical University, SanHao Street No. 36, HePing District, Shenyang, 110000, Liaoning, China
| | - Zhengwei Yuan
- Laboratory of Health Ministry for Congenital Malformation, ShengJing Hospital of China Medical University, SanHao Street No. 36, HePing District, Shenyang, 110000, Liaoning, China.
| | - Siyu Sun
- Department of Gastroenterology, ShengJing Hospital of China Medical University, SanHao Street No. 36, HePing District, Shenyang, 110000, Liaoning, China.
| | - Dongyan Liu
- Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, ShengJing Hospital of China Medical University, SanHao Street No. 36, HePing District, Shenyang, 110000, Liaoning, China.
| |
Collapse
|
66
|
An Y, Xu M, Yan M, Zhang H, Li C, Wang L, Liu C, Dong H, Chen L, Zhang L, Chen Y, Han X, Li Y, Wang D, Gao C. Erythrophagocytosis-induced ferroptosis contributes to pulmonary microvascular thrombosis and thrombotic vascular remodeling in pulmonary arterial hypertension. J Thromb Haemost 2025; 23:158-170. [PMID: 39357568 DOI: 10.1016/j.jtha.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/01/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Whether primary or just as a complication from the progression of pulmonary arterial hypertension (PAH), thrombosis seems to be an important player in this condition. The crosstalk between red blood cells (RBCs) and pulmonary microvascular endothelial cells (PMVECs) and their role in PAH remain undefined. OBJECTIVES The goals of this study were to assess the role of RBC-PMVEC interaction in microvascular thrombosis and thrombotic vascular remodeling under hypoxic conditions. METHODS We established an in vitro hypoxic coincubation model of RBC and PMVEC as well as a hypoxic mouse model. We investigated erythrophagocytosis (EP), ferroptosis, thrombosis tendency, and pulmonary hemodynamics in experimental PAH. RESULTS Increased EP in PMVEC triggered ferroptosis, enhanced procoagulant activity, and exacerbated vessel remodeling under hypoxic conditions. In the PAH mouse model induced by chronic hypoxia, EP-induced ferroptosis followed by upregulated TMEM16F led to a high tendency of thrombus formation and thrombotic vascular remodeling. Inhibition of ferroptosis or silencing of TMEM16F could alleviate hypercoagulable phenotype, reverse right ventricular systolic pressure, right ventricular hypertrophy index, and remodeling of pulmonary vessels. CONCLUSION These results illustrate the pathogenic RBC-PMVEC interactions in PAH. Inhibition EP, ferroptosis, or TMEM16F could be a novel therapeutic target to prevent PAH development and thrombotic complications.
Collapse
Affiliation(s)
- Yao An
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Minghui Xu
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Meishan Yan
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Hongyu Zhang
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Caixia Li
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Lifeng Wang
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Caixu Liu
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Haoran Dong
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Li Chen
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Lixin Zhang
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Yingli Chen
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Xu Han
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Yun Li
- Hematology Department, Daqing Oil Field General Hospital, Daqing, China
| | - Dongsheng Wang
- Department of Emergency, the Fifth Affiliated Hospital of Harbin Medical University, Daqing, China
| | - Chunyan Gao
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China.
| |
Collapse
|
67
|
Qin S, Zhu C, Chen C, Sheng Z, Cao Y. An emerging double‑edged sword role of ferroptosis in cardiovascular disease (Review). Int J Mol Med 2025; 55:16. [PMID: 39540363 PMCID: PMC11573318 DOI: 10.3892/ijmm.2024.5457] [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: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The pathophysiology of cardiovascular disease (CVD) is complex and presents a serious threat to human health. Cardiomyocyte loss serves a pivotal role in both the onset and progression of CVD. Among various forms of programmed cell death, ferroptosis, along with apoptosis, autophagy and pyroptosis, is closely linked to the advancement of CVD. Ferroptosis, a mechanism of cell death, is driven by the buildup of oxidized lipids and excess iron. This pathway is modulated by lipid, amino acid and iron metabolism. Key characteristics of ferroptosis include disrupted iron homeostasis, increased peroxidation of polyunsaturated fatty acids due to reactive oxygen species, decreased glutathione levels and inactivation of glutathione peroxidase 4. Treatments targeting ferroptosis could potentially prevent or alleviate CVD by inhibiting the ferroptosis pathway. Ferroptosis is integral to the pathogenesis of several types of CVD and inhibiting its occurrence in cardiomyocytes could be a promising therapeutic strategy for the future treatment of CVD. The present review provided an in‑depth analysis of advancements in understanding the mechanisms underlying ferroptosis. The present manuscript summarized the interplay between ferroptosis and CVDs, highlighting its dual roles in these conditions. Additionally, potential therapeutic targets within the ferroptosis pathway were discussed, alongside the current limitations and future directions of these novel treatment strategies. The present review may offer novel insights into preventive and therapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Sirun Qin
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Can Zhu
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Chenyang Chen
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhe Sheng
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yu Cao
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
68
|
Luan X, Chen P, Miao L, Yuan X, Yu C, Di G. Ferroptosis in organ ischemia-reperfusion injuries: recent advancements and strategies. Mol Cell Biochem 2025; 480:19-41. [PMID: 38556592 DOI: 10.1007/s11010-024-04978-2] [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: 11/14/2023] [Accepted: 02/24/2024] [Indexed: 04/02/2024]
Abstract
Ferroptosis is a newly discovered type of regulated cell death participated in multiple diseases. Different from other classical cell death programs such as necrosis and apoptosis, ferroptosis involving iron-catalyzed lipid peroxidation is characterized by Fe2+ accumulation and mitochondria alterations. The phenomenon of oxidative stress following organ ischemia-reperfusion (I/R) has recently garnered attention for its connection to the onset of ferroptosis and subsequent reperfusion injuries. This article provides a comprehensive overview underlying the mechanisms of ferroptosis, with a further focus on the latest research progress regarding interference with ferroptotic pathways in organ I/R injuries, such as intestine, lung, heart, kidney, liver, and brain. Understanding the links between ferroptosis and I/R injury may inform potential therapeutic strategies and targeted agents.
Collapse
Affiliation(s)
- Xiaoyu Luan
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Peng Chen
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
- Institute of Stem Cell and Regenerative Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Longyu Miao
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Xinying Yuan
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Chaoqun Yu
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Guohu Di
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China.
- Institute of Stem Cell and Regenerative Medicine, School of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
69
|
Zhang Y, Han R, Xu S, Shen B, Yu H, Chen J, Yao H, Huang S, Zhong Y. TMCO1 promotes ferroptosis and ECM deposition in glaucomatous trabecular meshwork via ERK1/2 signaling. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167530. [PMID: 39343416 DOI: 10.1016/j.bbadis.2024.167530] [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: 02/17/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Glaucoma, a leading cause of global blindness, is marked by irreversible retinal ganglion cells (RGCs) loss, elevated intraocular pressure (IOP), and extracellular matrix (ECM) deposition in the trabecular meshwork (TM). Transmembrane and coiled-coil domain protein 1 (TMCO1), implicated in calcium regulation, has potential links to primary open-angle glaucoma (POAG). Ferroptosis, an iron-dependent cell death mechanism driven by lipid peroxidation, is also observed in glaucoma. This study investigates the role of TMCO1 in POAG, focusing on its involvement in TM ECM deposition via ferroptosis induction and ERK1/2 phosphorylation inhibition. In both in vivo and in vitro models, we demonstrated that dexamethasone (DEX) stimulation upregulates TMCO1, leading to increased ECM deposition and ferroptosis in human trabecular meshwork cells (HTMCs). Furthermore, treatment with ferrostatin-1 (Fer-1), a ferroptosis inhibitor, significantly reduced ECM deposition and ferroptosis in HTMCs. These findings establish TMCO1 as a critical regulator of ferroptosis and ECM deposition through the ERK/MAPK pathway, positioning it as a promising therapeutic target for glaucoma.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, 197 Ruijin Er Road, 200025, Shanghai, China
| | - Ruiqi Han
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, 197 Ruijin Er Road, 200025, Shanghai, China
| | - Shushu Xu
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, 197 Ruijin Er Road, 200025, Shanghai, China
| | - Bingqiao Shen
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, 197 Ruijin Er Road, 200025, Shanghai, China; Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Huan Yu
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, 197 Ruijin Er Road, 200025, Shanghai, China
| | - Junjue Chen
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, 197 Ruijin Er Road, 200025, Shanghai, China
| | - Huiping Yao
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, 197 Ruijin Er Road, 200025, Shanghai, China.
| | - Shouyue Huang
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, 197 Ruijin Er Road, 200025, Shanghai, China.
| | - Yisheng Zhong
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, 197 Ruijin Er Road, 200025, Shanghai, China.
| |
Collapse
|
70
|
Archontakis-Barakakis P, Mavridis T, Chlorogiannis DD, Barakakis G, Laou E, Sessler DI, Gkiokas G, Chalkias A. Intestinal oxygen utilisation and cellular adaptation during intestinal ischaemia-reperfusion injury. Clin Transl Med 2025; 15:e70136. [PMID: 39724463 DOI: 10.1002/ctm2.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
The gastrointestinal tract can be deranged by ailments including sepsis, trauma and haemorrhage. Ischaemic injury provokes a common constellation of microscopic and macroscopic changes that, together with the paradoxical exacerbation of cellular dysfunction and death following restoration of blood flow, are collectively known as ischaemia-reperfusion injury (IRI). Although much of the gastrointestinal tract is normally hypoxemic, intestinal IRI results when there is inadequate oxygen availability due to poor supply (pathological hypoxia) or abnormal tissue oxygen use and metabolism (dysoxia). Intestinal oxygen uptake usually remains constant over a wide range of blood flows and pressures, with cellular function being substantively compromised when ischaemia leads to a >50% decline in intestinal oxygen consumption. Restoration of perfusion and oxygenation provokes additional injury, resulting in mucosal damage and disruption of intestinal barrier function. The primary cellular mechanism for sensing hypoxia and for activating a cascade of cellular responses to mitigate the injury is a family of heterodimer proteins called hypoxia-inducible factors (HIFs). The HIF system is connected to numerous biochemical and immunologic pathways induced by IRI and the concentration of those proteins increases during hypoxia and dysoxia. Activation of the HIF system leads to augmented transcription of specific genes in various types of affected cells, but may also augment apoptotic and inflammatory processes, thus aggravating gut injury. KEY POINTS: During intestinal ischaemia, mitochondrial oxygen uptake is reduced when cellular oxygen partial pressure decreases to below the threshold required to maintain normal oxidative metabolism. Upon reperfusion, intestinal hypoxia may persist because microcirculatory flow remains impaired and/or because available oxygen is consumed by enzymes, intestinal cells and neutrophils.
Collapse
Affiliation(s)
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital incorporating the National Children's Hospital (AMNCH), Dublin, Ireland
| | | | - Georgios Barakakis
- Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Laou
- Department of Anesthesiology, Agia Sophia Children's Hospital, Athens, Greece
| | - Daniel I Sessler
- Center for Outcomes Research and Department of Anesthesiology, UTHealth, Houston, Texas, USA
- Outcomes Research Consortium®, Houston, Texas, USA
| | - George Gkiokas
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Chalkias
- Outcomes Research Consortium®, Houston, Texas, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Critical Care Medicine, Tzaneio General Hospital, Piraeus, Greece
| |
Collapse
|
71
|
Li YC, Cheng ML. Carvedilol Confers Ferroptosis Resistance in HL-1 Cells by Upregulating GPX4, FTH1, and FTL1 and Inducing Metabolic Remodeling Under Hypoxia/Reoxygenation. Antioxidants (Basel) 2024; 14:7. [PMID: 39857341 PMCID: PMC11762394 DOI: 10.3390/antiox14010007] [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: 10/11/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Hypoxia/reoxygenation (HR) often occurs under cardiac pathological conditions, and HR-induced oxidative stress usually leads to cardiomyocyte damage. Carvedilol, a non-selective β-blocker, is used clinically to treat cardiac ischemia diseases. Moreover, Carvedilol has also been reported to have an antioxidant ability by reducing lipid peroxidation. However, the mechanism of Carvedilol to inhibit lipid peroxidation is still elusive. To explore the protective mechanism of Carvedilol to resist lipid peroxidation on cardiomyocytes, HL-1 cells were cultured under normoxia, hypoxia, and HR and treated with Carvedilol to investigate the alteration on metabolism, protein expression, and mRNA level to explain its oxidative mechanism. The study found that Carvedilol upregulated glutathione peroxidase 4 (GPX4) protein expression to resist HR-induced lipid peroxidation by metabolic remodeling under HR. Also, Carvedilol promoted ferroptosis-related genes, ferritin heavy chain 1 (FTH1) and ferritin light chain 1 (FTL1) mRNA levels, to reduce lipid peroxidation under both hypoxia and HR. In conclusion, our study explores a mechanism by which Carvedilol inhibits ferroptosis by upregulating GPX4, FTH1, and FTL1 levels to downregulate lipid peroxidation under HR. The study provides a potential strategy for using Carvedilol in clinical applications, inspiring further research and development in the area of heart diseases.
Collapse
Affiliation(s)
- Yi-Chin Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
| | - Mei-Ling Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City 33302, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| |
Collapse
|
72
|
Li Y, Liu C, Fang B, Chen X, Wang K, Xin H, Wang K, Yang SM. Ferroptosis, a therapeutic target for cardiovascular diseases, neurodegenerative diseases and cancer. J Transl Med 2024; 22:1137. [PMID: 39710702 DOI: 10.1186/s12967-024-05881-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The identification of ferroptosis represents a pivotal advancement in the field of cell death research, revealing an entirely novel mechanism of cellular demise and offering new insights into the initiation, progression, and therapeutic management of various diseases. Ferroptosis is predominantly induced by intracellular iron accumulation, lipid peroxidation, or impairments in the antioxidant defense system, culminating in membrane rupture and consequent cell death. Studies have associated ferroptosis with a wide range of diseases, and by enhancing our comprehension of its underlying mechanisms, we can formulate innovative therapeutic strategies, thereby providing renewed hope for patients.
Collapse
Affiliation(s)
- Yinghui Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Cuiyun Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bo Fang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xinzhe Chen
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Kai Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China.
| | - Kun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Su-Min Yang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
73
|
Zou P, He Q, Xia H, Zhong W. Ferroptosis and its impact on common diseases. PeerJ 2024; 12:e18708. [PMID: 39713140 PMCID: PMC11663406 DOI: 10.7717/peerj.18708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/23/2024] [Indexed: 12/24/2024] Open
Abstract
Ferroptosis is a novel form of programmed cell death characterized by iron accumulation, lipid peroxidation, and a decline in antioxidant capacity, all of which are regulated by gene expression. The onset of numerous diseases is closely associated with ferroptosis. Common diseases affect a large population, reduce the quality of life, and impose an increased burden on the healthcare system. The role of ferroptosis in common diseases, its therapeutic potential, and even its translation into clinical drug treatments are currently significant research topics worldwide. This study preliminarily explores the theoretical basis of ferroptosis, its mechanism and treatment prospect in common diseases including ischaemia-reperfusion injury, inflammatory bowel diseases, liver fibrosis, acute kidney injury, diabetic kidney disease, stroke, Alzheimer's disease, cardiovascular disease, immune and cancer. This review provides a theoretical foundation for the further study and development of ferroptosis, as well as for the prevention and treatment of common diseases.
Collapse
Affiliation(s)
- Pengjian Zou
- Department of Pediatric Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qiuming He
- Department of Pediatric Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Zhong
- Department of Pediatric Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
74
|
Liu X, Wang W, Zhu F, Xu H, Ge G, Liang X, Yang H, Xu Y, Xu W, Wei M, Zhou Q, Geng D. Osteoblastic ferroptosis inhibition by small-molecule promoting GPX4 activation for peri-prosthetic osteolysis therapy. J Nanobiotechnology 2024; 22:758. [PMID: 39696565 DOI: 10.1186/s12951-024-03049-4] [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: 09/27/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Peri-prosthesis osteolysis (PPO) represents the most severe complication of total joint arthroplasty (TJA) surgery and imposes the primary cause of prosthesis failure and subsequent revision surgery. Antiresorptive therapies are usually prescribed to treat PPO, especially for elderly people. Nevertheless, the efficacy of anti-osteoporotic medications remains constrained. Recent therapeutic strategies to promote periprosthetic osseointegration by restoring osteoblast function are considered more effective approaches. However, the precise mechanism underlying the inhibition of osteogenesis triggered by wear particles remains enigmatic. Herein, we demonstrate that wear particles inhibit osteoblast function by inducing ferroptosis to sabotage extracellular mineralization and arouse periprosthetic osteolysis. The suppression of ferroptosis could significantly rescue osteogenesis thus alleviating PPO. Furthermore, Glutathione Peroxidase 4 (GPX4) has been identified as a key target in regulating osteoblastic ferroptosis. By utilizing virtual screening techniques, we have successfully conducted a comprehensive screening of a natural compound known as Urolithin A (UA), which exhibits remarkable inhibition of osteoblastic ferroptosis while simultaneously promoting the process of osteogenesis through its precise targeting mechanism on GPX4. Meanwhile, UA improves the osteolytic conditions significantly in vivo even when the adjunction of titanium (Ti) nanoparticles. This strategy has great potential in treating peri-prosthesis osteolysis and potentially broadens the scope of clinical therapy.
Collapse
Affiliation(s)
- Xin Liu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Wei Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Feng Zhu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Haibo Xu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Gaoran Ge
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xiaolong Liang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Huilin Yang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yaozeng Xu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Wei Xu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| | - Minggang Wei
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| | - Qi Zhou
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, 200070, China.
| | - Dechun Geng
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China.
| |
Collapse
|
75
|
Tan R, Ge C, Yan Y, Guo H, Han X, Zhu Q, Du Q. Deciphering ferroptosis in critical care: mechanisms, consequences, and therapeutic opportunities. Front Immunol 2024; 15:1511015. [PMID: 39737174 PMCID: PMC11682965 DOI: 10.3389/fimmu.2024.1511015] [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: 10/14/2024] [Accepted: 12/03/2024] [Indexed: 01/01/2025] Open
Abstract
Ischemia-reperfusion injuries (IRI) across various organs and tissues, along with sepsis, significantly contribute to the progression of critical illnesses. These conditions disrupt the balance of inflammatory mediators and signaling pathways, resulting in impaired physiological functions in human tissues and organs. Ferroptosis, a distinct form of programmed cell death, plays a pivotal role in regulating tissue damage and modulating inflammatory responses, thereby influencing the onset and progression of severe illnesses. Recent studies highlight that pharmacological agents targeting ferroptosis-related proteins can effectively mitigate oxidative stress caused by IRI in multiple organs, alleviating associated symptoms. This manuscript delves into the mechanisms and signaling pathways underlying ferroptosis, its role in critical illnesses, and its therapeutic potential in mitigating disease progression. We aim to offer a novel perspective for advancing clinical treatments for critical illnesses.
Collapse
Affiliation(s)
- Ruimin Tan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Chen Ge
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yating Yan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - He Guo
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xumin Han
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiong Zhu
- Department of Orthopaedics, The People’s Hospital Of Shizhu, Chongqing, China
| | - Quansheng Du
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
76
|
Liu Y, Miao R, Xia J, Zhou Y, Yao J, Shao S. Infection of Helicobacter pylori contributes to the progression of gastric cancer through ferroptosis. Cell Death Discov 2024; 10:485. [PMID: 39622791 PMCID: PMC11612470 DOI: 10.1038/s41420-024-02253-3] [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: 09/25/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
Helicobacter pylori (H. pylori) is a gram-negative pathogen that colonizes gastric epithelial cells, and its chronic infection is the primary risk factor for the development of gastric cancer (GC). Ferroptosis is an iron-dependent form of cell death characterized by intracellular lipid peroxide accumulation and reactive oxygen species (ROS) imbalance. There is evidence suggesting that pathogens can manipulate ferroptosis to facilitate their replication, transmission, and pathogenesis. However, the interaction between ferroptosis and H. pylori infection requires further elucidation. We reviewed the mechanism of ferroptosis and found that H. pylori virulence factors such as cytotoxin-associated gene A (CagA), vacuolating cytotoxin A (VacA), neutrophil-activating protein A (NapA), superoxide dismutase B (SodB), γ-glutamyl transpeptidase (gGT), lipopolysaccharide (LPS), and outer inflammatory protein A (OipA) affected glutathione (GSH), ROS, and lipid oxidation to regulate ferroptosis. It also affected the progression of GC by regulating ferroptosis-related indicators through abnormal gene expression after H. pylori infected gastric mucosa cells. Finally, we discuss the potential application value of ferroptosis inducers, inhibitors and other drugs in treating H. pylori-infected GC patients while acknowledging that their interactions are still not fully understood.
Collapse
Affiliation(s)
- Yun Liu
- Department of Gastroenterology, Institute of Digestive Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Renjie Miao
- Department of Clinical laboratory, Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jinxuan Xia
- Zhenjiang Mental Health Center, Jiangsu, China
| | - Yong Zhou
- Department of Gastroenterology, Institute of Digestive Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Jun Yao
- Department of Gastroenterology, Institute of Digestive Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Shihe Shao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
77
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [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: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
78
|
Jia D, Wu K, Luo J, Xu X, Pan W, Zhao M, Li S, Gong J, Gong J. Wogonin Alleviates DCD Liver Ischemia/Reperfusion Injury by Regulating ALOX15/iNOS-mediated Ferroptosis. Transplantation 2024; 108:2374-2385. [PMID: 38946036 DOI: 10.1097/tp.0000000000005123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
BACKGROUND Donation after circulatory death livers are more susceptible to ischemia/reperfusion injury (IRI) because of a longer period of warm ischemia. Growing evidence now suggests that ferroptosis plays a key regulatory role in the development of IRI, so targeting ferroptosis may be an effective strategy to alleviate IRI in liver transplantation (LT). METHODS Using donation after circulatory death LT models in rats and oxygen-glucose deprivation/reoxygenation (OGD/R) models in BRL-3A cells, we tested the effect of the Chinese medicine monomer wogonin on liver IRI and explored the specific mechanism. RESULTS Wogonin attenuated liver IRI and increased the survival rate of rats by inhibiting lipid peroxidation and ferroptosis. Mechanistically, arachidonic acid 15-lipoxygenase-1 (ALOX15) and inducible nitric oxide synthase (iNOS) were identified as potential targets of baicalein through bioinformatics analysis combined with in vivo and in vitro experiments. This result was further confirmed by molecular docking and cellular thermal shift assays. Finally, we silenced ALOX15 and iNOS in the OGD/R cell model and found that silencing ALOX15 and iNOS could reproduce the regulatory effect of wogonin and abrogate the regulatory effect of wogonin. CONCLUSIONS In brief, this study emphasizes that wogonin exerts a protective effect in liver IRI through the regulation of ALOX15- and iNOS-mediated ferroptosis. ALOX15 and iNOS are potential targets for intervention in IRI induced by LT, and wogonin is a drug candidate for LT patients.
Collapse
Affiliation(s)
- Degong Jia
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Hou M, Li X, Chen F, Tan Z, Han X, Liu J, Zhou J, Shi Y, Zhang J, Lv J, Leng Y. Naringenin alleviates intestinal ischemia/reperfusion injury by inhibiting ferroptosis via targeting YAP/STAT3 signaling axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156095. [PMID: 39383632 DOI: 10.1016/j.phymed.2024.156095] [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: 04/29/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Intestinal ischemia/reperfusion injury (IRI) is a significant clinical emergency, and investigating novel therapeutic approaches and understanding their underlying mechanisms is essential for improving patient outcomes. Naringenin (Nar), a flavanone present in tomatoes and citrus fruits, is frequently consumed in the human diet and recognized for having immunomodulatory, anti-inflammatory, and antioxidant properties. Despite Nar being able to alleviate intestinal IRI, the exact molecular mechanisms remain elusive. PURPOSE To investigate Nar's protective properties on intestinal IRI and elucidate the mechanisms, a comprehensive approach that combines network pharmacology analysis with experimental verification in vitro and in vivo was adopted. METHODS The oxygen-glucose deprivation/reoxygenation (OGD/R) model in IEC-6 cells and a murine model of intestinal IRI were used. Nar's effects on intestinal IRI were assessed through histological analysis using H&E staining and tight junction (TJ) protein expression. Ferroptosis-related parameters, including iron levels, superoxide dismutase (SOD), glutathione (GSH), reactive oxygen species (ROS), malondialdehyde (MDA), and mitochondrial morphology, were analyzed. Network pharmacology was utilized to predict the pathways through which Nar exerts its anti-ferroptosis effects. Further mechanistic insights were obtained through si-RNA transfection, YAP inhibitor (verteporfin, VP) treatment, ferroptosis inhibitor (Ferrostatin-1) and ferroptosis inducer (Erastin) application, co-immunoprecipitation (Co-IP) and Western blotting. RESULTS Our results revealed that pretreatment with Nar significantly mitigated intestinal tissue damage and improved gut barrier function, as evidenced by increased TJ proteins (ZO-1 and Occludin). Nar reduced iron, MDA, and ROS, while it increased GSH and SOD levels. Additionally, Nar alleviated mitochondrial damage in mice. Nar treatment increased GPX4 and SLC7A11, while decreasing ACSL4 levels both in vivo and in vitro. Network pharmacology analysis suggested that Nar may target the Hippo signaling pathway. Notably, YAP, a key transcriptional co-activator within the Hippo pathway, was downregulated in intestinal IRI mice and OGD/R-induced IEC-6 cells. Nar pretreatment activated YAP, thereby augmenting anti-ferroptosis effects. The inhibition of YAP activation by VP or YAP knockdown increased p-STAT3 expression, thereby diminishing Nar's efficacy. Co-IP and immunofluorescence studies confirmed the interaction between YAP and STAT3. CONCLUSION This study shows that Nar can inhibit ferroptosis in intestinal IRI via activating YAP, which in turn suppresses STAT3 phosphorylation, thereby unveiling a novel mechanism and supporting Nar's potential to be a promising therapeutic agent for intestinal IRI.
Collapse
Affiliation(s)
- Min Hou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Xiaoxi Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Feng Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Zhiguo Tan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Xiaoxia Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Jie Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Jia Zhou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Yajing Shi
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Jianmin Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Jipeng Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| | - Yufang Leng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China; Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
80
|
Fan Q, Chang H, Tian L, Zheng B, Liu R, Li Z. Methane saline suppresses ferroptosis via the Nrf2/HO-1 signaling pathway to ameliorate intestinal ischemia-reperfusion injury. Redox Rep 2024; 29:2373657. [PMID: 39023011 PMCID: PMC11259071 DOI: 10.1080/13510002.2024.2373657] [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: 07/20/2024] Open
Abstract
OBJECTIVES Intestinal ischemia-reperfusion (I/R) injury is a multifactorial and complex clinical pathophysiological process. Current research indicates that the pathogenesis of intestinal I/R injury involves various mechanisms, including ferroptosis. Methane saline (MS) has been demonstrated to primarily exert anti-inflammatory and antioxidant effects in I/R injury. In this study, we mainly investigated the effect of MS on ferroptosis in intestinal I/R injury and determined its potential mechanism. METHODS In vivo and in vitro intestinal I/R injury models were established to validate the relationship between ferroptosis and intestinal I/R injury. MS treatment was applied to assess its impact on intestinal epithelial cell damage, intestinal barrier disruption, and ferroptosis. RESULTS MS treatment led to a reduction in I/R-induced intestinal epithelial cell damage and intestinal barrier disruption. Moreover, similar to treatment with ferroptosis inhibitors, MS treatment reduced ferroptosis in I/R, as indicated by a decrease in the levels of intracellular pro-ferroptosis factors, an increase in the levels of anti-ferroptosis factors, and alleviation of mitochondrial damage. Additionally, the expression of Nrf2/HO-1 was significantly increased after MS treatment. However, the intestinal protective and ferroptosis inhibitory effects of MS were diminished after the use of M385 to inhibit Nrf2 in mice or si-Nrf2 in Caco-2 cells. DISCUSSION We proved that intestinal I/R injury was mitigated by MS and that the underlying mechanism involved modulating the Nrf2/HO-1 signaling pathway to decrease ferroptosis. MS could be a promising treatment for intestinal I/R injury.
Collapse
Affiliation(s)
- Qingrui Fan
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
- Xi’an Medical University, Xi’an, People’s Republic of China
| | - Hulin Chang
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
- Department of Hepatobiliary Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Lifei Tian
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Bobo Zheng
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Ruiting Liu
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Zeyu Li
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| |
Collapse
|
81
|
Zeng Q, Jiang T. Molecular mechanisms of ferroptosis in cardiovascular disease. Mol Cell Biochem 2024; 479:3181-3193. [PMID: 38374233 DOI: 10.1007/s11010-024-04940-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/12/2024] [Indexed: 02/21/2024]
Abstract
Ferroptosis is a newly recognized type of regulated cell death that is characterized by the accumulation of iron and lipid peroxides in cells. Studies have shown that ferroptosis plays a significant role in the pathogenesis of various diseases, including cardiovascular diseases. In cardiovascular disease, ferroptosis is associated with ischemia-reperfusion injury, myocardial infarction, heart failure, and atherosclerosis. The molecular mechanisms underlying ferroptosis include the iron-dependent accumulation of lipid peroxidation products, glutathione depletion, and dysregulation of lipid metabolism, among others. This review aims to summarize the current knowledge of the molecular mechanisms of ferroptosis in cardiovascular disease and discuss the potential therapeutic strategies targeting ferroptosis as a treatment for cardiovascular disease.
Collapse
Affiliation(s)
- Qun Zeng
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Tingting Jiang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| |
Collapse
|
82
|
Feng Y, Shi M, Zhang Y, Li X, Yan L, Xu J, Liu C, Li M, Bai F, Yuan F, Sun Y, Liu R, Zhao Y, Yang L, Zhang Y, Guo Y, Zhang J, Zhou R, Liu P. Protocatechuic acid relieves ferroptosis in hepatic lipotoxicity and steatosis via regulating NRF2 signaling pathway. Cell Biol Toxicol 2024; 40:104. [PMID: 39589556 PMCID: PMC11599353 DOI: 10.1007/s10565-024-09953-7] [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: 06/26/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Ferroptosis represents a newly programmed cell death, and the process is usually accompanied with iron-dependent lipid peroxidation. Importantly, ferroptosis is implicated in a myriad of diseases. Recent literature suggests a potential position of ferroptosis in the pathogenesis of metabolic dysfunction-associated fatty liver disease (MAFLD), the most widespread liver ailment worldwide. Intriguingly, several functional genes and metabolic pathways central to ferroptosis are regulated by nuclear factor erythroid-derived 2-like 2 (NRF2). In current work, we aim to identify protocatechuic acid (PCA), a primary metabolite of antioxidant polyphenols, as a potent NRF2 activator and ferroptosis inhibitor in the hepatic lipotoxicity and steatosis models. Herein, both NRF2+/+ and NRF2-/- cell lines and mice were used to analyze the importance of NRF2 in PCA function, and hepatic lipotoxicity and steatosis models were induced by palmitic acid and high-fat diet respectively. Our results indicated that ferroptosis was mitigated by PCA intervention in hepatic cells. Furthermore, PCA exhibited therapeutic efficacy against ferroptosis, as well as hepatic lipotoxicity and steatosis. The protective role of PCA was predominantly mediated through NRF2 activation, potentially elucidating a pivotal mechanism underlying PCA's therapeutic impact on MAFLD. Additionally, the augmented mitochondrial TCA cycle activity observed in hepatic lipotoxicity and steatosis models was ameliorated by PCA, in part via NRF2-dependent pathways, further bolstering PCA's anti-ferroptosis properties. Collectively, our findings underscore PCA's potential in alleviating hepatic ferroptosis, lipotoxicity and steatosis via inducing activation of NRF2 signaling pathway, offering a promising strategy for the therapy of MAFLD as well as related lipid metabolic disorders.
Collapse
Affiliation(s)
- Yetong Feng
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengjiao Shi
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Yi Zhang
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Xinyan Li
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Liangwen Yan
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Jiayi Xu
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Chenyue Liu
- Department of Medical Image, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Miaomiao Li
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Fengyun Bai
- Shaanxi Dongtai Pharmaceutical Co., LTD, Xianyang, China
| | - Fenyue Yuan
- Shaanxi Dongtai Pharmaceutical Co., LTD, Xianyang, China
| | - Ying Sun
- Shaanxi Dongtai Pharmaceutical Co., LTD, Xianyang, China
| | - Rongrong Liu
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Yaping Zhao
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Lan Yang
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yinggang Zhang
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Ying Guo
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Jian Zhang
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Rui Zhou
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Pengfei Liu
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China.
- Key Laboratory of Environment and Genes Related To Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
83
|
Zeng X, Jiang S, Wu Y, Zhong L, Liu X. Regulatory role of AMPK/Nrf2 signaling pathway in sevoflurane-enhanced intestinal protection against ischemia-reperfusion injury. Int J Biochem Cell Biol 2024; 177:106702. [PMID: 39581489 DOI: 10.1016/j.biocel.2024.106702] [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: 08/12/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Intestinal ischemia-reperfusion (I/R) injury is common in clinical settings and is associated with high mortality. Sevoflurane, a widely used anesthetic, has long recognized for its protective effects against intestinal I/R injury, though the underlying mechanisms remain largely uncharacterized. In this study, using both in vivo and in vitro models, we uncovered a novel role of sevoflurane in preventing ferroptotic cell death during intestinal I/R injury. Sevoflurane treatment activated transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) and upregulated its target genes involved in iron sequestration (FTL) and glutathione biosynthesis (SLC7A11 and GCLM). These changes reduced intracellular ferrous iron levels and alleviated iron-dependent oxidative stress and lipid peroxidation, a hallmark of ferroptosis. Importantly, through large-scale kinome screening, we revealed that sevoflurane-induced Nrf2 activation was mediated by AMP-activated protein kinase (AMPK). Sevoflurane treatment activated AMPK, which subsequently phosphorylated Nrf2 and prevented its degradation. Stabilized Nrf2 then entered nucleus, where it promoted the transcription of downstream targets. We concluded that sevoflurane exerts anti-ferroptoic function in intestinal I/R through the AMPK/Nrf2 signaling pathway. These results expand our knowledge about the pathogenesis of intestinal I/R injury, and provide novel insights for optimizing clinical treatments and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaohua Zeng
- Anesthesiology Department, General Hospital of the Yang Tze River Shipping, Wuhan Brain Hospital, Wuhan, China
| | - Shan Jiang
- Anesthesiology Department, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinghui Wu
- Anesthesiology Department, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Zhong
- Anesthesiology Department, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Liu
- Anesthesiology Department, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
84
|
Liu J, Ren J, Zhou L, Tan K, Du D, Xu L, Cao W, Zhang Y. Proteomic and lipidomic analysis of the mechanism underlying astragaloside IV in mitigating ferroptosis through hypoxia-inducible factor 1α/heme oxygenase 1 pathway in renal tubular epithelial cells in diabetic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118517. [PMID: 38972525 DOI: 10.1016/j.jep.2024.118517] [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: 04/05/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The limitations of modern medicine in mitigating the pathological process of diabetic kidney disease (DKD) necessitate novel, precise, and effective prevention and treatment methods. Huangqi, the root of Astragalus membranaceus Fisch. ex Bunge has been used in traditional Chinese medicine for various kidney ailments. Astragaloside IV (AS-IV), the primary pharmacologically active compound in A. membranaceus, is involved in lipid metabolism regulation; however, its potential in ameliorating renal damage in DKD remains unexplored. AIM OF THE STUDY To elucidate the specific mechanism by which AS-IV moderates DKD progression. MATERIALS AND METHODS A murine model of DKD and high glucose-induced HK-2 cells were treated with AS-IV. Furthermore, multiomics analysis, molecular docking, and molecular dynamics simulations were performed to elucidate the mechanism of action of AS-IV in DKD, which was validated using molecular biological methods. RESULTS AS-IV regulated glucose and lipid metabolism in DKD, thereby mitigating lipid deposition in the kidneys. Proteomic analysis identified 12 proteins associated with lipid metabolism regulated by AS-IV in the DKD renal tissue. Additionally, lipid metabolomic analysis revealed that AS-IV upregulated and downregulated 4 beneficial and 79 harmful lipid metabolites, respectively. Multiomics analysis further indicated a positive correlation between the top-ranked differential protein heme oxygenase (HMOX)1 and the levels of various harmful lipid metabolites and a negative correlation with the levels of beneficial lipid metabolites. Furthermore, enrichment of both ferroptosis and hypoxia-inducible factor (HIF)-1 signaling pathways during the AS-IV treatment of DKD was observed using proteomic analysis. Validation results showed that AS-IV effectively reduced ferroptosis in DKD-affected renal tubular epithelial cells by inhibiting HIF-1α/HMOX1 pathway activity, upregulating glutathione peroxidase-4 and ferritin heavy chain-1 expression, and downregulating acyl-CoA synthetase long-chain family member-4 and transferrin receptor-1 expression. Our findings demonstrate the potential of AS-IV in mitigating DKD pathology by downregulating the HIF-1α/HMOX1 signaling pathway, thereby averting ferroptosis in renal tubular epithelial cells. CONCLUSIONS AS-IV is a promising treatment strategy for DKD via the inhibition of ferroptosis in renal tubular epithelial cells. The findings of this study may help facilitate the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Jun Liu
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1., Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, PR China.
| | - Jing Ren
- College of Traditional Chinese Medicine, Chongqing Medical and Pharmaceutical College, No. 82, University Town Middle Road, Shapingba District, Chongqing, 401331, PR China.
| | - Linlan Zhou
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1., Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, PR China.
| | - Kaiyue Tan
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1, Medical College Road, Yuzhong District, Chongqing, 400016, PR China; Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, 400016, PR China.
| | - Donglin Du
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, PR China.
| | - Lei Xu
- Laboratory Animal Center, Chongqing Medical University, No. 1, Medical College Road, Yuzhong District, Chongqing, 400016, PR China.
| | - Wenfu Cao
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1., Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, PR China; College of Traditional Chinese Medicine, Chongqing Medical University, No. 1, Medical College Road, Yuzhong District, Chongqing, 400016, PR China; Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, 400016, PR China.
| | - Yudi Zhang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1, Medical College Road, Yuzhong District, Chongqing, 400016, PR China; Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, 400016, PR China; College of Combination of Chinese and Western Medicine, Chongqing College of Traditional Chinese Medicine, No. 61, Puguobao Road, Bicheng Street, Bishan District, Chongqing, 402760, PR China.
| |
Collapse
|
85
|
You Y, Qian Z, Jiang Y, Chen L, Wu D, Liu L, Zhang F, Ning X, Zhang Y, Xiao J. Insights into the pathogenesis of gestational and hepatic diseases: the impact of ferroptosis. Front Cell Dev Biol 2024; 12:1482838. [PMID: 39600338 PMCID: PMC11588751 DOI: 10.3389/fcell.2024.1482838] [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: 08/20/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Ferroptosis, a distinct form of non-apoptotic cell death characterized by iron dependency and lipid peroxidation, is increasingly linked to various pathological conditions in pregnancy and liver diseases. It plays a critical role throughout pregnancy, influencing processes such as embryogenesis, implantation, and the maintenance of gestation. A growing body of evidence indicates that disruptions in these processes can precipitate pregnancy-related disorders, including pre-eclampsia (PE), gestational diabetes mellitus (GDM), and intrahepatic cholestasis of pregnancy (ICP). Notably, while ICP is primarily associated with elevated maternal serum bile acid levels, its precise etiology remains elusive. Oxidative stress induced by bile acid accumulation is believed to be a significant factor in ICP pathogenesis. Similarly, the liver's susceptibility to oxidative damage underscores the importance of lipid metabolism dysregulation and impaired iron homeostasis in the progression of liver diseases such as alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), cholestatic liver injury, autoimmune hepatitis (AIH), acute liver injury, viral hepatitis, liver fibrosis, and hepatocellular carcinoma (HCC). This review discusses the shared signaling mechanisms of ferroptosis in gestational and hepatic diseases, and explores recent advances in understanding the mechanisms of ferroptosis and its potential role in the pathogenesis of gestational and hepatic disorders, with the aim of identifying viable therapeutic targets.
Collapse
Affiliation(s)
- Yilan You
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Zhiwen Qian
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Ying Jiang
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Lingyan Chen
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Danping Wu
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Lu Liu
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Feng Zhang
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Xin Ning
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Yan Zhang
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Jianping Xiao
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| |
Collapse
|
86
|
Liu H, Yue Q, Zhang W, Ding Q, Yang J, Lin M, Sun J. Xinglou Chengqi Decoction Protects against Cerebral Ischemia/Reperfusion Injury by Inhibiting Ferroptosis via SLC7A11/GPX4 Signaling. Adv Biol (Weinh) 2024; 8:e2400180. [PMID: 39334526 DOI: 10.1002/adbi.202400180] [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: 04/02/2024] [Revised: 08/27/2024] [Indexed: 09/30/2024]
Abstract
Xinglou Chengqi decoction (XLCQD) is a Chinese formula that offers benefits in ischemic stroke. However, the underlying mechanism of the effects of XLCQD-mediated anti-ischemic stroke effects remains obscure. This study investigates the ferroptosis mechanism of XLCQD against cerebral ischemia/reperfusion (I/R) injury using rat models of middle cerebral artery occlusion/reperfusion (MCAO/R). Ferroptosis differs from traditional cell death pathways and is linked to oxidative stress-induced lipid peroxidation and glutathione (GSH) depletion, which is essential to the development of ischemic stroke. In this study, it is shown that XLCQD improves brain infarction, neurological dysfunction, and histopathological changes caused by MCAO/R exposure, and improving I/R-induced oxidative damage through inhibition of ferroptosis via (Solute Carrier Family 7 Member 11) SLC7A11/ (glutathione peroxidase 4) GPX4 pathway. Interestingly, it is found that XLCQD-mediated protection in I/R is reversed by the silence of SLC7A11. XLCQD intervention significantly promotes GSH content and suppresses Reactive Oxygen Species(ROS), iron accumulation, as well as Malondialdehyde (MDA) generation, are markedly abrogated when SLC7A11 is knockdown by SLC7A11-shRNA transfection, indicating that SLC7A11 is the main target of XLCQD to further trigger intracellular events. In conclusion, XLCQD attenuates in vivo cerebral I/R injury by reducing ferroptosis via the SLC7A11/GPX4 pathway.
Collapse
Affiliation(s)
- Hua Liu
- Department of Neurology, Yixing Traditional Chinese Medicine Hospital, Yixing, Jiangsu, 214200, China
| | - Qiyu Yue
- Division of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wenyue Zhang
- Department of Neurology, Yixing Traditional Chinese Medicine Hospital, Yixing, Jiangsu, 214200, China
| | - Qi Ding
- Department of Rehabilitation, Yixing Traditional Chinese Medicine Hospital, Yixing, Jiangsu, 214200, China
| | - Junjie Yang
- Yixing Traditional Chinese Medicine Hospital, Yixing, Jiangsu, 214200, China
| | - Mu Lin
- Yixing Traditional Chinese Medicine Hospital, Yixing, Jiangsu, 214200, China
| | - Jia Sun
- Department of Neurology, Yixing Traditional Chinese Medicine Hospital, Yixing, Jiangsu, 214200, China
| |
Collapse
|
87
|
Yang Z, Gao W, Yang K, Chen W, Chen Y. The protective role of RACK1 in hepatic ischemia‒reperfusion injury-induced ferroptosis. Inflamm Res 2024; 73:1961-1979. [PMID: 39292271 DOI: 10.1007/s00011-024-01944-y] [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: 02/06/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
Although ferroptosis plays a crucial role in hepatic ischemia‒reperfusion injury (IRI), the molecular mechanisms underlying this process remain unclear. We aimed to explore the potential involvement of the receptor for activated C kinase 1 (RACK1) in hepatic IRI-triggered ferroptosis. Using hepatocyte-specific RACK1 knockout mice and alpha mouse liver 12 (AML12) cells, we conducted a series of in vivo and in vitro experiments. We found that RACK1 has a protective effect on hepatic IRI-induced ferroptosis. Specifically, RACK1 was found to interact with AMPKα through its 1-93 amino acid (aa) region, which facilitates the phosphorylation of AMPKα at threonine 172 (Thr172), ultimately exerting an antiferroptotic effect. Furthermore, the long noncoding RNA (lncRNA) ZNFX1 Antisense 1 (ZFAS1) directly binds to aa 181-317 of RACK1. ZFAS1 has a dual impact on RACK1 by promoting its ubiquitin‒proteasome-mediated degradation and inhibiting its expression at the transcriptional level, which indirectly exacerbates hepatic IRI-induced ferroptosis. These findings underscore the protective role of RACK1 in hepatic IRI-induced ferroptosis and showcase its potential as a prophylactic target for hepatic IRI mitigation.
Collapse
Affiliation(s)
- Zelong Yang
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Wenjie Gao
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Kai Yang
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Weigang Chen
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yong Chen
- Department of Hepatobiliary Surgery, Xi Jing Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
88
|
Huang M, Cheng S, Li Z, Chen J, Wang C, Li J, Zheng H. Preconditioning Exercise Inhibits Neuron Ferroptosis and Ameliorates Brain Ischemia Damage by Skeletal Muscle-Derived Exosomes via Regulating miR-484/ACSL4 Axis. Antioxid Redox Signal 2024; 41:769-792. [PMID: 38545792 DOI: 10.1089/ars.2023.0492] [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: 04/25/2024]
Abstract
Aims: Although there is evidence that patients with stroke who exercise regularly before stroke have a better prognosis than those who do not exercise, the detailed mechanism remains unclear. Moreover, neuronal death plays a central role in neurological dysfunction caused by ischemic stroke. Thus, we investigated whether exercise could reduce stroke-induced neuronal death and its associated mediators in the current study. Results: Ferroptosis was the most dominant form of programmed cell death in neurons. Preconditioning exercise before stroke improved the neurological function and decreased the infarct area in rats with ischemic stroke. Preconditioning exercise attenuated stroke-induced ferroptosis by reducing lipid peroxidation (LPO) production, upregulating glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11), and downregulating acyl-CoA synthetase long-chain family member 4 (ACSL4). High-throughput sequencing and dual luciferase reporter assays revealed that exercise-induced exosomal miR-484 inhibits Acsl4 expression. Moreover, we showed that exercise-induced exosomal miR-484 is mainly derived from skeletal muscle, and the neuroprotective effect of preconditioning exercise is suppressed by inhibiting miR-484 production in skeletal muscle. Innovation: This study suggested that neuronal ferroptosis is the most dominant form of programmed cell death in a hypoxic environment. Moreover, we showed that the ferroptosis pathway is a potential therapeutic target in ischemic stroke and that preconditioning exercise could be an effective antioxidant intervention for cerebral ischemia. Conclusion: Our work revealed that preconditioning exercise before stroke exerts neuroprotective effects against brain ischemia by skeletal muscle-derived exosomal miR-484 via inhibiting ferroptosis. Antioxid. Redox Signal. 41, 769-792.
Collapse
Affiliation(s)
- Mudan Huang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shimei Cheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ziwen Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jinshuo Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chuangjia Wang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jun Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
89
|
Guo Y, Han Y, Zhang J, Zhou Y, Wei M, Yu L. Identification and Experimental Validation of Prognostic miRNA Signature and Ferroptosis-Related Key Genes in Cervical Squamous Cell Carcinoma. Cancer Med 2024; 13:e70415. [PMID: 39526479 PMCID: PMC11551785 DOI: 10.1002/cam4.70415] [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: 12/07/2023] [Revised: 10/05/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVES This study aimed to investigate the prognostic value of miRNAs and ferroptosis-related genes in cervical squamous cell carcinoma. METHODS We mined data from public databases for differentially expressed miRNAs, ferroptosis-related genes, and clinical parameters and constructed a prognostic risk model. The predictive performance of the model was evaluated using survival and receiver operating characteristic curve analyses. We combined the clinicopathological features to construct a nomogram and evaluated its efficacy using calibration and clinical decision curves. The correlation between miRNA characteristics, risk score, and the tumor microenvironment was also studied. Next, consensus and key genes were screened, and their biological functions were analyzed using KEGG, GO, GSEA, and drug sensitivity analysis. Finally, the expression of miRNAs and key genes was detected using qRT-PCR and western blotting to verify the prediction results. RESULTS Seven miRNA signatures (miR-100-3p, miR-301a-5p, miR-331-3p, miR-425-5p, miR-502-3p, miR-505-5p, and miR-629-3p) were generated, and prognostic risk and nomogram models were successfully constructed. These models exhibited good accuracy. miRNA signatures correlated with the tumor microenvironment. Twelve consensus genes and three key genes (SLC2A1, ANO6, and TXNIP) were screened and their biofunctional diversity was identified using various analytical methods. qRT-PCR and western blotting were used to verify the expression of miR-301a-5p, miR-505-5p, SLC2A1, and TXNIP in cervical squamous carcinoma. The results were consistent with those of bioinformatics analyses. CONCLUSIONS Seven miRNAs may serve as prognostic biomarkers of cervical squamous cell carcinoma. SLC2A1, ANO6, and TXNIP are associated with cervical squamous cell carcinoma and may serve as ferroptosis-related markers of the disease.
Collapse
Affiliation(s)
- Yan Guo
- Department of GynecologyShanxi Medical University First HospitalTaiyuanChina
| | - Yana Han
- Department of GynecologyShanxi Medical University First HospitalTaiyuanChina
| | - Junjie Zhang
- Department of NeurosurgeryShanxi Medical University Second HospitalTaiyuanChina
| | - Yanbin Zhou
- Department of Teaching Affairs SectionShanxi Medical University First HospitalTaiyuanChina
| | - Meiyan Wei
- Department of GynecologyShanxi Medical University First HospitalTaiyuanChina
| | - Lijun Yu
- Department of GynecologyShanxi Medical University First HospitalTaiyuanChina
| |
Collapse
|
90
|
Kambey PA, Wu J, Liu W, Su M, Buberwa W, Tang C. Targeting serum response factor (SRF) deactivates ΔFosB and mitigates Levodopa-induced dyskinesia in a mouse model of Parkinson's disease. Gene Ther 2024; 31:614-624. [PMID: 39384937 DOI: 10.1038/s41434-024-00492-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA) is currently the preferred treatment for Parkinson's Disease (PD) and is considered the gold standard. However, prolonged use of L-DOPA in patients can result in involuntary movements known as Levodopa-induced dyskinesia (LID), which includes uncontrollable dystonia affecting the trunk, limbs, and face. The role of ΔFosB protein, a truncated splice variant of the FosB gene, in LID has been acknowledged, but its underlying mechanism has remained elusive. Here, using a mouse model of Parkinson's disease treated with chronic levodopa we demonstrate that serum response factor (SRF) binds to the FosB promoter, thereby activating FosB expression and levodopa induced-dyskinetic movements. Western blot analysis demonstrates a significant increase in SRF expression in the dyskinetic group compared to the control group. Knocking down SRF significantly reduced abnormal involuntary movements (AIMS) and ΔFosB expression compared to the control. Conversely, overexpression of SRF led to an increase in ΔFosB expression and worsened levodopa-induced dyskinesia. To shed light on the regulatory role of the Akt signaling pathway in this phenomenon, we administered the Akt agonist SC79 to PD mouse models via intraperitoneal injection, followed by L-DOPA administration. The expression of SRF, ΔFosB, and phosphorylated Akt (p-Akt) significantly increased in this group compared to the group receiving normal saline to signify that these happen through Akt signaling pathway. Collectively, our findings identify a promising therapeutic target for addressing levodopa-induced dyskinesia.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
- Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Huangpu District, Guangzhou, China.
| | - Jiao Wu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - WenYa Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Mingyu Su
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Wokuheleza Buberwa
- Department of Neurology, The second affiliated hospital of Xi'an Jiaotong University, 710049, Xi'an, China
| | - Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
91
|
Zhu J, Wu Y, Zhang L, Bai B, Han W, Wang H, Mei Q. Epithelial Piezo1 deletion ameliorates intestinal barrier damage by regulating ferroptosis in ulcerative colitis. Free Radic Biol Med 2024; 224:272-286. [PMID: 39216559 DOI: 10.1016/j.freeradbiomed.2024.08.039] [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: 06/28/2024] [Revised: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Ferroptosis, a recently discovered form of regulated cell death, has been implicated in the development of ulcerative colitis (UC). While Piezo1's role in inducing ferroptosis in chondrocytes and pulmonary endothelial cells is documented, its regulatory function in ferroptosis and intestinal epithelial cells in UC remains unclear. To address this, colonic tissue samples from patients with UC were examined, and specific intestinal epithelial Piezo1-deficient (Piezo1ΔIEC) mice were created to investigate Piezo1's role in UC pathogenesis. Elevated epithelial Piezo1 levels were observed in patients with UC, correlating with increased ferroptosis and tight junction (TJ) disruption. In dextran sulfate sodium (DSS)-induced colitis, Piezo1ΔIEC mice exhibited significantly reduced intestinal inflammation and improved gut barrier function compared to wild-type (WT) mice. Moreover, Piezo1 deficiency in colitis mice and lipopolysaccharide (LPS)-stimulated Caco-2 cells led to higher TJ protein levels, reduced lipid peroxidation, enhanced mitochondrial function, and altered expression of ferroptosis-associated proteins. Additionally, erastin, a ferroptosis activator, reversed the protective effect of Piezo1 silencing against LPS-induced ferroptosis in Caco-2 cells. Mechanistically, Piezo1 was found to regulate ferroptosis via the AMPK/mTOR signaling pathway. These findings highlight a novel role for Piezo1 deletion in mitigating ferroptosis in intestinal epithelial cells, suggesting Piezo1 as a potential therapeutic target for UC treatment.
Collapse
Affiliation(s)
- Jiejie Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Yumei Wu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Luyao Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Bingqing Bai
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Wei Han
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.
| | - Qiao Mei
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China.
| |
Collapse
|
92
|
Tian L, Liu Q, Guo H, Zang H, Li Y. Fighting ischemia-reperfusion injury: Focusing on mitochondria-derived ferroptosis. Mitochondrion 2024; 79:101974. [PMID: 39461581 DOI: 10.1016/j.mito.2024.101974] [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: 07/04/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of mortality and morbidity. Current treatments for IRI have limited efficacy and novel therapeutic strategies are needed. Mitochondrial dysfunction not only initiates IRI but also plays a significant role in ferroptosis pathogenesis. Recent studies have highlighted that targeting mitochondrial pathways is a promising therapeutic approach for ferroptosis-induced IRI. The association between ferroptosis and IRI has been reviewed many times, but our review provides the first comprehensive overview with a focus on recent mitochondrial research. First, we present the role of mitochondria in ferroptosis. Then, we summarize the evidence on mitochondrial manipulation of ferroptosis in IRI and review recent therapeutic strategies aimed at targeting mitochondria-related ferroptosis to mitigate IRI. We hope our review will provide new ideas for the treatment of IRI and accelerate the transition from bench to bedside.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, China
| | - Hong Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Honggang Zang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
93
|
Wang S, Qin M, Fan X, Jiang C, Hou Q, Ye Z, Zhang X, Yang Y, Xiao J, Wallace K, Rastegar-Kashkooli Y, Peng Q, Jin D, Wang J, Wang M, Ding R, Tao J, Kim YT, Bhawal UK, Wang J, Chen X, Wang J. The role of metal ions in stroke: Current evidence and future perspectives. Ageing Res Rev 2024; 101:102498. [PMID: 39243890 DOI: 10.1016/j.arr.2024.102498] [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: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Metal ions play a pivotal role in maintaining optimal brain function within the human body. Nevertheless, the accumulation of these ions can result in irregularities that lead to brain damage and dysfunction. Disruptions of metal ion homeostasis can result in various pathologies, including inflammation, redox dysregulation, and blood-brain barrier disruption. While research on metal ions has chiefly focused on neurodegenerative diseases, little attention has been given to their involvement in the onset and progression of stroke. Recent studies have identified cuproptosis and confirmed ferroptosis as significant factors in stroke pathology, underscoring the importance of metal ions in stroke pathology, including abnormal ion transport, neurotoxicity, blood-brain barrier damage, and cell death. Additionally, it provides an overview of contemporary metal ion chelators and detection techniques, which may offer novel approaches to stroke treatment.
Collapse
Affiliation(s)
- Shaoshuai Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Non-commissioned Officer School of Army Medical University, Shijiazhuang, Hebei 050000, China
| | - Mengzhe Qin
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Qingchuan Hou
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ziyi Ye
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinru Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yunfan Yang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jingyu Xiao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kevin Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qinfeng Peng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Dongqi Jin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruoqi Ding
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jin Tao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ujjal K Bhawal
- Center for Global Health Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India; Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba 271-8587, Japan
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
94
|
Zhan M, Liu D, Yao L, Wang W, Zhang R, Xu Y, Wang Z, Yan Q, Fang Q, Du J, Chen L. Gas6/AXL Alleviates Hepatic Ischemia/Reperfusion Injury by Inhibiting Ferroptosis via the PI3K/AKT Pathway. Transplantation 2024; 108:e357-e369. [PMID: 38725107 PMCID: PMC11495534 DOI: 10.1097/tp.0000000000005036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 10/24/2024]
Abstract
BACKGROUND Hepatic ischemia/reperfusion (I/R) injury is a major cause of complications in clinical liver surgery. AXL receptor tyrosine kinase (AXL) is a member of the TAM receptor tyrosine kinase family (TYRO3, AXL, and MERTK). Our previous study has shown that AXL expression was markedly upregulated in liver transplantation patients. However, the underlying mechanism of AXL in hepatic I/R injury remains unclear. METHODS A mouse liver warm I/R model and a primary hepatocyte hypoxia/reoxygenation model were established to investigate the role of AXL activation and ferroptosis in hepatic I/R injury by pretreating with recombinant mouse growth arrest-specific protein 6 (AXL activator) or R428 (AXL inhibitor). Moreover, we used LY294002 (phosphatidylinositol 3-kinase [PI3K] inhibitor) to evaluate the relationship between the PI3K/AKT (the Ser and Thr kinase AKT) pathway and ferroptosis in hepatic I/R injury. RESULTS Hepatic I/R injury decreased phosphorylation AXL expression and enhanced ferroptosis in liver transplantation patients and hepatic I/R-subjected mice. AXL activation attenuated lipid peroxidation and ferroptosis in hepatic I/R injury in vivo and in vitro. Inhibition of AXL activation exacerbated liver pathological damage and liver dysfunction, as well as iron accumulation and lipid peroxidation in hepatic I/R injury. Mechanistically, activated growth arrest-specific protein 6/AXL and its downstream PI3K/AKT signaling pathway inhibited ferroptosis during hepatic I/R injury. CONCLUSIONS AXL activation protects against hepatic I/R injury by preventing ferroptosis through the PI3K/AKT pathway. This study is the first investigation on the AXL receptor and ferroptosis, and activating AXL to mitigate ferroptosis may be an innovative therapeutic strategy to combat hepatic I/R injury.
Collapse
Affiliation(s)
- Mengting Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Deng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Lei Yao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Weizhi Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Ruixin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Yaru Xu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Zhen Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Anesthesiology, Ningbo First Hospital, Ningbo, China
| | - Qi Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qi Fang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jian Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Infectious Disease Research Center, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lijian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| |
Collapse
|
95
|
Zeng L, Jin X, Xiao QA, Jiang W, Han S, Chao J, Zhang D, Xia X, Wang D. Ferroptosis: action and mechanism of chemical/drug-induced liver injury. Drug Chem Toxicol 2024; 47:1300-1311. [PMID: 38148561 DOI: 10.1080/01480545.2023.2295230] [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: 06/09/2023] [Revised: 09/12/2023] [Accepted: 11/28/2023] [Indexed: 12/28/2023]
Abstract
Drug-induced liver injury (DILI) is characterized by hepatocyte injury, cholestasis injury, and mixed injury. The liver transplantation is required for serious clinical outcomes such as acute liver failure. Current studies have found that many mechanisms were involved in DILI, such as mitochondrial oxidative stress, apoptosis, necroptosis, autophagy, ferroptosis, etc. Ferroptosis occurs when hepatocytes die from iron-dependent lipid peroxidation and plays a key role in DILI. After entry into the liver, where some drugs or chemicals are metabolized, they convert into hepatotoxic substances, consume reduced glutathione (GSH), and decrease the reductive capacity of GSH-dependent GPX4, leading to redox imbalance in hepatocytes and increase of reactive oxygen species (ROS) and lipid peroxidation level, leading to the undermining of hepatocytes; some drugs facilitated the autophagy of ferritin, orchestrating the increased ion level and ferroptosis. The purpose of this review is to summarize the role of ferroptosis in chemical- or drug-induced liver injury (chemical/DILI) and how natural products inhibit ferroptosis to prevent chemical/DILI.
Collapse
Affiliation(s)
- Li Zeng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Xueli Jin
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Qing-Ao Xiao
- Department of Interventional Radiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Wei Jiang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Shanshan Han
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Jin Chao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Ding Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Xuan Xia
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Department of Physiology and Pathophysiology, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Decheng Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| |
Collapse
|
96
|
Li Z, Fu J, Jiang K, Gao J, Guo Y, Li C, Zhao L, Nam J, Gao H. Hyperbaric Oxygen Improves Cognitive Impairment Induced by Hypoxia via Upregulating the Expression of Oleic Acid and MBOAT2 of Mice. Antioxidants (Basel) 2024; 13:1320. [PMID: 39594462 PMCID: PMC11591255 DOI: 10.3390/antiox13111320] [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: 09/27/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
Cognitive impairment (CI) causes severe impairment of brain function and quality of life of patients, which brings a great burden to society. Cerebral hypoxia is an important factor in the pathogenesis of CI. Hyperbaric oxygen (HBO) therapy may mitigate hypoxia-induced CI, but its efficacy and mechanisms are not fully understood. In this study, a mice model of CI induced by hypoxia environment was established, then behavioral tests, pathological examination, metabolomic and lipidomic analyses, and molecular biology were used to assess the impact of HBO on hypoxia-induced CI. HBO was found to alleviate CI and pathological damage of hypoxia mice. Metabolomic, lipidomic, and molecular biology analyses showed that HBO increased the levels of oleic acid (OA) and membrane-bound O-acyltransferase 2 (MBOAT2), thereby altering the composition of membrane phospholipids (PLs) and reducing hypoxia-induced neuronal ferroptosis (FPT) to interfere with cognitive function in mice. In vitro experiments confirmed that OA and MBOAT2 led to membrane PL remodeling in a mutually dependent manner, affecting cell resistance to hypoxia-FPT. The results emphasized the combined effect value of OA and MBOAT2 in HBO for hypoxia-induced CI, and provided a novel perspective for the treatment of CI by HBO.
Collapse
Affiliation(s)
- Zhen Li
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jun Fu
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
- Innocation Academy of Testing Technology, Research and Experiment Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Kaiyuan Jiang
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Jie Gao
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Yuejun Guo
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Chen Li
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Liangcai Zhao
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hongchang Gao
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, Wenzhou Medical University, Wenzhou 325035, China (J.F.); (K.J.); (C.L.); (L.Z.)
- Innocation Academy of Testing Technology, Research and Experiment Center, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
97
|
Sun M, Li Q, Zou Z, Liu J, Gu Z, Li L. The mechanisms behind heatstroke-induced intestinal damage. Cell Death Discov 2024; 10:455. [PMID: 39468029 PMCID: PMC11519599 DOI: 10.1038/s41420-024-02210-0] [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: 04/15/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
With the frequent occurrence of heatwaves, heatstroke (HS) is expected to become one of the main causes of global death. Being a multi-organized disease, HS can result in circulatory disturbance and systemic inflammatory response, with the gastrointestinal tract being one of the primary organs affected. Intestinal damage plays an initiating and promoting role in HS. Multiple pathways result in damage to the integrity of the intestinal epithelial barrier due to heat stress and hypoxia brought on by blood distribution. This usually leads to intestinal leakage as well as the infiltration and metastasis of toxins and pathogenic bacteria in the intestinal cavity, which will eventually cause inflammation in the whole body. A large number of studies have shown that intestinal damage after HS involves the body's stress response, disruption of oxidative balance, disorder of tight junction proteins, massive cell death, and microbial imbalance. Based on these damage mechanisms, protecting the intestinal barrier and regulating the body's inflammatory and immune responses are effective treatment strategies. To better understand the pathophysiology of this complex process, this review aims to outline the potential processes and possible therapeutic strategies for intestinal damage after HS in recent years.
Collapse
Affiliation(s)
- Minshu Sun
- Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qin Li
- Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhimin Zou
- Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Liu
- Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengtao Gu
- Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Li Li
- Department of Intensive Care Unit, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
98
|
He S, Bai J, Zhang L, Yuan H, Ma C, Wang X, Guan X, Mei J, Zhu X, Xin W, Zhu D. Superenhancer-driven circRNA Myst4 involves in pulmonary artery smooth muscle cell ferroptosis in pulmonary hypertension. iScience 2024; 27:110900. [PMID: 39351203 PMCID: PMC11440257 DOI: 10.1016/j.isci.2024.110900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/19/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
The abnormal expression of circular RNAs (circRNAs) is emerging as a critical cause in regulation of pathological changes of hypoxic pulmonary hypertension (PH), in which ferroptosis is a new pathological change reported recently. However, how circRNAs regulate ferroptosis remains unclear. Here, we proved a significant decrease in circMyst4 expression in hypoxia. In vitro assays revealed that circMyst4 alleviated hypoxic pulmonary artery smooth muscle cell (PASMC) ferroptosis through directly combing with DDX5 in the nucleus to promote GPX4 mRNA processing and inhibiting the formation of the Eef1a1/ACSL4 complex in the cytoplasm. Additionally, superenhancer (SE) was verified to drive the generation of circMyst4. In vivo assays revealed that circMyst4 inhibited the progression of hypoxic PH. Overall, SE-driven circMyst4 may be a new potential therapeutic target for mediating PASMC ferroptosis through promoting DDX5-regulated GPX4 mRNA processing and inhibiting the binding between Eef1a1 and ACSL4.
Collapse
Affiliation(s)
- Siyu He
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
| | - June Bai
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
| | - Lixin Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Hao Yuan
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
| | - Cui Ma
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Xiaoying Wang
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Xiaoyu Guan
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
| | - Jian Mei
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Xiangrui Zhu
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Wei Xin
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200031, P.R. China
| | - Daling Zhu
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
- Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, Harbin Medical University, Harbin 150081, P.R. China
| |
Collapse
|
99
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-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: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
100
|
Han J, Zheng D, Liu PS, Wang S, Xie X. Peroxisomal homeostasis in metabolic diseases and its implication in ferroptosis. Cell Commun Signal 2024; 22:475. [PMID: 39367496 PMCID: PMC11451054 DOI: 10.1186/s12964-024-01862-w] [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: 07/31/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024] Open
Abstract
Peroxisomes are dynamic organelles involved in various cellular processes, including lipid metabolism, redox homeostasis, and intracellular metabolite transfer. Accumulating evidence suggests that peroxisomal homeostasis plays a crucial role in human health and disease, particularly in metabolic disorders and ferroptosis. The abundance and function of peroxisomes are regulated by a complex interplay between biogenesis and degradation pathways, involving peroxins, membrane proteins, and pexophagy. Peroxisome-dependent lipid metabolism, especially the synthesis of ether-linked phospholipids, has been implicated in modulating cellular susceptibility to ferroptosis, a newly discovered form of iron-dependent cell death. This review discusses the current understanding of peroxisome homeostasis, its roles in redox regulation and lipid metabolism, and its implications in human diseases. We also summarize the main mechanisms of ferroptosis and highlight recent discoveries on how peroxisome-dependent metabolism and signaling influence ferroptosis sensitivity. A better understanding of the interplay between peroxisomal homeostasis and ferroptosis may provide new insights into disease pathogenesis and reveal novel therapeutic strategies for peroxisome-related metabolic disorders and ferroptosis-associated diseases.
Collapse
Affiliation(s)
- Jiwei Han
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China
| | - Daheng Zheng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China
| | - Pu-Ste Liu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Shanshan Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangdong, China
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China.
| |
Collapse
|