101
|
Huang Q, Tian H, Tian L, Zhao X, Li L, Zhang Y, Qiu Z, Lei S, Xia Z. Inhibiting Rev-erbα-mediated ferroptosis alleviates susceptibility to myocardial ischemia-reperfusion injury in type 2 diabetes. Free Radic Biol Med 2023; 209:135-150. [PMID: 37805047 DOI: 10.1016/j.freeradbiomed.2023.09.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/09/2023]
Abstract
The complex progression of type-2 diabetes (T2DM) may result in increased susceptibility to myocardial ischemia-reperfusion (IR) injury. IR injuries in multiple organs involves ferroptosis. Recently, the clock gene Rev-erbα has aroused considerable interest as a novel therapeutic target for metabolic and ischemic heart diseases. Herein, we investigated the roles of Rev-erbα and ferroptosis in myocardial IR injury during T2DM and its potential mechanisms. A T2DM model, myocardial IR and a tissue-specific Rev-erbα-/- mouse in vivo were established, and a high-fat high glucose environment with hypoxia-reoxygenation (HFHG/HR) in H9c2 were also performed. After myocardial IR, glycolipid profiles, creatine kinase-MB, AI, and the expression of Rev-erbα and ferroptosis-related proteins were increased in diabetic rats with impaired cardiac function compared to non-diabetic rats, regardless of the time at which IR was induced. The ferroptosis inhibitor ferrostatin-1 decreased AI in diabetic rats given IR and LPO levels in cells treated with HFHG/HR, as well as the expression of Rev-erbα and ACSL4. The ferroptosis inducer erastin increased AI and LPO levels and ACSL4 expression. Treatment with the circadian regulator nobiletin and genetically targeting Rev-erbα via siRNA or CRISPR/Cas9 technology both protected against severe myocardial injury and decreased Rev-erbα and ACSL4 expression, compared to the respective controls. Taken together, these data suggest that ferroptosis is involved in the susceptibility to myocardial IR injury during T2DM, and that targeting Rev-erbα could alleviate myocardial IR injury by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Qin Huang
- Department of Anaesthesiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei, PR China
| | - Hao Tian
- Department of Anaesthesiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei, PR China
| | - Liqun Tian
- Department of Anaesthesiology, The First Affiliated Hospital of Chongqing Medical University, PR China
| | - Xiaoshuai Zhao
- Department of Anaesthesiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei, PR China
| | - Lu Li
- Department of Anaesthesiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei, PR China
| | - Yuxi Zhang
- Department of Anaesthesiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei, PR China
| | - Zhen Qiu
- Department of Anaesthesiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei, PR China
| | - Shaoqing Lei
- Department of Anaesthesiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei, PR China
| | - Zhongyuan Xia
- Department of Anaesthesiology, Wuhan Univ, Renmin Hospital, Wuhan 430060, Hubei, PR China.
| |
Collapse
|
102
|
Shan Y, Guan C, Wang J, Qi W, Chen A, Liu S. Impact of ferroptosis on preeclampsia: A review. Biomed Pharmacother 2023; 167:115466. [PMID: 37729725 DOI: 10.1016/j.biopha.2023.115466] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
Preeclampsia (PE) is usually associated with the accumulation of reactive oxygen species (ROS) resulting from heightened oxidative stress (OS). Ferroptosis is a unique type of lipid peroxidation-induced iron-dependent cell death distinct from traditional apoptosis, necroptosis, and pyroptosis and most likely contributes considerable to PE pathogenesis. At approximately 10-12 weeks of gestation, trophoblasts create an environment rich in oxygen and iron. In patients with PE, ferroptosis-related genes such as HIF1 and MAPK8 are downregulated, whereas PLIN2 is upregulated. Furthermore, miR-30b-5p overexpression inhibits solute carrier family 11 member 2, resulting in a decrease in glutathione levels and an increase in the labile iron pool. At the maternal-fetal interface, physiological hypoxia/reperfusion and excessive iron result in lipid peroxidation and ROS production. Owing to the high expression of Fpn and polyunsaturated fatty acid-containing phospholipid-related enzymes, including acyl-CoA synthetase long-chain family member 4, lysophosphatidylcholine acyl-transferase 3, and spermidine/spermine N1-acetyltransferase 1, trophoblasts become more susceptible to OS and ROS damage. In stage 1, the injured trophoblasts exhibit poor invasion and incomplete uterine spiral artery remodeling caused by ferroptosis, leading to placental ischemia and hypoxia. Subsequently, ferroptosis marked by OS occurs in stage 2, eventually causing PE. We aimed to explore the new therapeutic target of PE through OS in ferroptosis.
Collapse
Affiliation(s)
- Yuping Shan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chengcheng Guan
- Laboratory Department, Qingdao Haici Hospital, Qingdao, China
| | - Jingli Wang
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weihong Qi
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aiping Chen
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Shiguo Liu
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
103
|
Hao M, Jiang Y, Zhang Y, Yang X, Han J. Ferroptosis regulation by methylation in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188972. [PMID: 37634887 DOI: 10.1016/j.bbcan.2023.188972] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/31/2023] [Accepted: 08/20/2023] [Indexed: 08/29/2023]
Abstract
Epigenetic regulation plays a critical role in cancer development and progression. Methylation is an important epigenetic modification that influences gene expression by adding a methyl group to nucleic acids and proteins. Ferroptosis is a new form of regulated cell death triggered by the accumulation of iron and lipid peroxidation. Emerging evidence have shown that methylation regulation plays a significant role in the regulation of ferroptosis in cancer. This review aims to explore the methylation regulation of ferroptosis in cancer, including reactive oxygen species and iron bio-logical activity, amino acid and lipid metabolism, and drugs interaction. The findings of this review may provide new insights and strategies for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Mengqiu Hao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China
| | - Yixin Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China
| | - Yang Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China; Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuyang Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China; Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Junhong Han
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, PR China.
| |
Collapse
|
104
|
Huang Y, Li X, Zhang Z, Xiong L, Wang Y, Wen Y. Photodynamic Therapy Combined with Ferroptosis Is a Synergistic Antitumor Therapy Strategy. Cancers (Basel) 2023; 15:5043. [PMID: 37894410 PMCID: PMC10604985 DOI: 10.3390/cancers15205043] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/20/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Ferroptosis is a programmed death mode that regulates redox homeostasis in cells, and recent studies suggest that it is a promising mode of tumor cell death. Ferroptosis is regulated by iron metabolism, lipid metabolism, and intracellular reducing substances, which is the mechanism basis of its combination with photodynamic therapy (PDT). PDT generates reactive oxygen species (ROS) and 1O2 through type I and type II photochemical reactions, and subsequently induces ferroptosis through the Fenton reaction and the peroxidation of cell membrane lipids. PDT kills tumor cells by generating excessive cytotoxic ROS. Due to the limited laser depth and photosensitizer enrichment, the systemic treatment effect of PDT is not good. Combining PDT with ferroptosis can compensate for these shortcomings. Nanoparticles constructed by photosensitizers and ferroptosis agonists are widely used in the field of combination therapy, and their targeting and biological safety can be improved through modification. These nanoparticles not only directly kill tumor cells but also further exert the synergistic effect of PDT and ferroptosis by activating antitumor immunity, improving the hypoxia microenvironment, and inhibiting the tumor angiogenesis. Ferroptosis-agonist-induced chemotherapy and PDT-induced ablation also have good clinical application prospects. In this review, we summarize the current research progress on PDT and ferroptosis and how PDT and ferroptosis promote each other.
Collapse
Affiliation(s)
- Yunpeng Huang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Xiaoyu Li
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China;
| | - Zijian Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Li Xiong
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Yongxiang Wang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Yu Wen
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| |
Collapse
|
105
|
Li Y, Ma JQ, Wang CC, Zhou J, Sun YD, Wei XL, Zhao ZQ. Ferroptosis: A potential target of macrophages in plaque vulnerability. Open Life Sci 2023; 18:20220722. [PMID: 37791060 PMCID: PMC10543703 DOI: 10.1515/biol-2022-0722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/04/2023] [Accepted: 08/16/2023] [Indexed: 10/05/2023] Open
Abstract
Plaque vulnerability has been the subject of several recent studies aimed at reducing the risk of stroke and carotid artery stenosis. Atherosclerotic plaque development is a complex process involving inflammation mediated by macrophages. Plaques become more vulnerable when the equilibrium between macrophage recruitment and clearance is disturbed. Lipoperoxides, which are affected by iron levels in cells, are responsible for the cell death seen in ferroptosis. Ferroptosis results from lipoperoxide-induced mitochondrial membrane toxicity. Atherosclerosis in ApoE(-/-) mice is reduced when ferroptosis is inhibited and iron intake is limited. Single-cell sequencing revealed that a ferroptosis-related gene was substantially expressed in atherosclerosis-modeled macrophages. Since ferroptosis can be regulated, it offers hope as a non-invasive method of treating carotid plaque. In this study, we discuss the role of ferroptosis in atherosclerotic plaque vulnerability, including its mechanism, regulation, and potential future research directions.
Collapse
Affiliation(s)
- Yu Li
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Ji-Qing Ma
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Chao-Chen Wang
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Jian Zhou
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Yu-Dong Sun
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University,
Nanjing201411, China
| | - Xiao-Long Wei
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Zhi-Qing Zhao
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| |
Collapse
|
106
|
Mu M, Zhang Q, Zhao C, Li X, Chen Z, Sun X, Yu J. 3-Bromopyruvate overcomes cetuximab resistance in human colorectal cancer cells by inducing autophagy-dependent ferroptosis. Cancer Gene Ther 2023; 30:1414-1425. [PMID: 37558749 PMCID: PMC10581902 DOI: 10.1038/s41417-023-00648-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related death worldwide. Cetuximab, in combination with chemotherapy, is effective for treating patients with wild-type KRAS/BRAF metastatic CRC (mCRC). However, intrinsic or acquired drug resistance often limits the use of cetuximab. In this study, we investigated the potential of co-treatment with 3-Bromopyruvate (3-BP) and cetuximab to overcome cetuximab resistance in CRC, both in vitro and in vivo. Our results demonstrated that the co-treatment of 3-BP and cetuximab synergistically induced an antiproliferative effect in both CRC cell lines with intrinsic cetuximab resistance (DLD-1 (KRASG13D/-) and HT29 (BRAFV600E)) and in a cetuximab-resistant cell line derived from Caco-2 with acquired resistance (Caco-2-CR). Further analysis revealed that co-treatment induced ferroptosis, autophagy, and apoptosis. Mechanistically, co-treatment inhibited FOXO3a phosphorylation and degradation and activated the FOXO3a/AMPKα/pBeclin1 and FOXO3a/PUMA pathways, leading to the promotion of ferroptosis, autophagy, and apoptosis in DLD-1 (KRASG13D/-), HT29 (BRAFV600E), and Caco-2-CR cells. In conclusion, our findings suggest that co-treatment with 3-BP and cetuximab could be a promising strategy to overcome cetuximab resistance in human CRC.
Collapse
Affiliation(s)
- Mingchao Mu
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Qin Zhang
- Department of Dermatology, Northwest Hospital, the Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, Shaanxi, China
| | - Chenye Zhao
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Xiaopeng Li
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Zilu Chen
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Xuejun Sun
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China.
| | - Junhui Yu
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China.
| |
Collapse
|
107
|
Hu S, Chu Y, Zhou X, Wang X. Recent advances of ferroptosis in tumor: From biological function to clinical application. Biomed Pharmacother 2023; 166:115419. [PMID: 37666176 DOI: 10.1016/j.biopha.2023.115419] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/21/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023] Open
Abstract
Ferroptosis is a recently recognized form of cell death with distinct features in terms of morphology, biochemistry, and molecular mechanisms. Unlike other types of cell death, ferroptosis is characterized by iron dependence, reactive oxygen species accumulation and lipid peroxidation. Recent studies have demonstrated that selective autophagy plays a vital role in the induction of ferroptosis, including ferritinophagy, lipophagy, clockophagy, and chaperone-mediated autophagy. Emerging evidence has indicated the involvement of ferroptosis in tumorigenesis through regulating various biological processes, including tumor growth, metastasis, stemness, drug resistance, and recurrence. Clinical and preclinical studies have found that novel therapies targeting ferroptosis exert great potential in the treatment of tumors. This review provides a comprehensive overview of the molecular mechanisms in ferroptosis, especially in autophagy-driven ferroptosis, discusses the recent advances in the biological roles of ferroptosis in tumorigenesis, and highlights the application of novel ferroptosis-targeted therapies in the clinical treatment of tumors.
Collapse
Affiliation(s)
- Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yurou Chu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| |
Collapse
|
108
|
Zhang H, Sun C, Sun Q, Li Y, Zhou C, Sun C. Susceptibility of acute myeloid leukemia cells to ferroptosis and evasion strategies. Front Mol Biosci 2023; 10:1275774. [PMID: 37818101 PMCID: PMC10561097 DOI: 10.3389/fmolb.2023.1275774] [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: 08/10/2023] [Accepted: 09/15/2023] [Indexed: 10/12/2023] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematologic malignancy with a 5-year survival rate of less than 30%. Continuous updating of diagnostic and therapeutic strategies has not been effective in improving the clinical benefit of AML. AML cells are prone to iron metabolism imbalance due to their unique pathological characteristics, and ferroptosis is a novel cell death mode that is dominated by three cellular biological processes: iron metabolism, oxidative stress and lipid metabolism. An in-depth exploration of the unique ferroptosis mechanism in AML can provide new insights for the diagnosis and treatment of this disease. This study summarizes recent studies on ferroptosis in AML cells and suggests that the metabolic characteristics, gene mutation patterns, and dependence on mitochondria of AML cells greatly increase their susceptibility to ferroptosis. In addition, this study suggests that AML cells can establish a variety of strategies to evade ferroptosis to maintain their survival during the process of occurrence and development, and summarizes the related drugs targeting ferroptosis pathway in AML treatment, which provides development directions for the subsequent mechanism research and clinical treatment of AML.
Collapse
Affiliation(s)
- Hanyun Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunjie Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qi Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ye Li
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Chao Zhou
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| |
Collapse
|
109
|
Sun S, Shen J, Jiang J, Wang F, Min J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct Target Ther 2023; 8:372. [PMID: 37735472 PMCID: PMC10514338 DOI: 10.1038/s41392-023-01606-1] [Citation(s) in RCA: 188] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 09/23/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated cell death with distinct characteristics, including altered iron homeostasis, reduced defense against oxidative stress, and abnormal lipid peroxidation. Recent studies have provided compelling evidence supporting the notion that ferroptosis plays a key pathogenic role in many diseases such as various cancer types, neurodegenerative disease, diseases involving tissue and/or organ injury, and inflammatory and infectious diseases. Although the precise regulatory networks that underlie ferroptosis are largely unknown, particularly with respect to the initiation and progression of various diseases, ferroptosis is recognized as a bona fide target for the further development of treatment and prevention strategies. Over the past decade, considerable progress has been made in developing pharmacological agonists and antagonists for the treatment of these ferroptosis-related conditions. Here, we provide a detailed overview of our current knowledge regarding ferroptosis, its pathological roles, and its regulation during disease progression. Focusing on the use of chemical tools that target ferroptosis in preclinical studies, we also summarize recent advances in targeting ferroptosis across the growing spectrum of ferroptosis-associated pathogenic conditions. Finally, we discuss new challenges and opportunities for targeting ferroptosis as a potential strategy for treating ferroptosis-related diseases.
Collapse
Affiliation(s)
- Shumin Sun
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Shen
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Jiang
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
110
|
Pourcelot E, El Samra G, Mossuz P, Moulis JM. Molecular Insight into Iron Homeostasis of Acute Myeloid Leukemia Blasts. Int J Mol Sci 2023; 24:14307. [PMID: 37762610 PMCID: PMC10531764 DOI: 10.3390/ijms241814307] [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: 08/20/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Acute myeloid leukemia (AML) remains a disease of gloomy prognosis despite intense efforts to understand its molecular foundations and to find efficient treatments. In search of new characteristic features of AML blasts, we first examined experimental conditions supporting the amplification of hematological CD34+ progenitors ex vivo. Both AML blasts and healthy progenitors heavily depended on iron availability. However, even if known features, such as easier engagement in the cell cycle and amplification factor by healthy progenitors, were observed, multiplying progenitors in a fully defined medium is not readily obtained without modifying their cellular characteristics. As such, we measured selected molecular data including mRNA, proteins, and activities right after isolation. Leukemic blasts showed clear signs of metabolic and signaling shifts as already known, and we provide unprecedented data emphasizing disturbed cellular iron homeostasis in these blasts. The combined quantitative data relative to the latter pathway allowed us to stratify the studied patients in two sets with different iron status. This categorization is likely to impact the efficiency of several therapeutic strategies targeting cellular iron handling that may be applied to eradicate AML blasts.
Collapse
Affiliation(s)
- Emmanuel Pourcelot
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), University Grenoble Alpes, INSERM U1055, 38000 Grenoble, France; (E.P.); (G.E.S.)
- Department of Biological Hematology, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043 Grenoble, CEDEX a9, France;
| | - Ghina El Samra
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), University Grenoble Alpes, INSERM U1055, 38000 Grenoble, France; (E.P.); (G.E.S.)
| | - Pascal Mossuz
- Department of Biological Hematology, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043 Grenoble, CEDEX a9, France;
- Team “Epigenetic and Cellular Signaling”, Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700 Grenoble, France
| | - Jean-Marc Moulis
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), University Grenoble Alpes, INSERM U1055, 38000 Grenoble, France; (E.P.); (G.E.S.)
- University Grenoble Alpes, CEA, IRIG, 38000 Grenoble, France
| |
Collapse
|
111
|
Xie Y, Zhou Y, Wang J, Du L, Ren Y, Liu F. Ferroptosis, autophagy, tumor and immunity. Heliyon 2023; 9:e19799. [PMID: 37810047 PMCID: PMC10559173 DOI: 10.1016/j.heliyon.2023.e19799] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/20/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Ferroptosis was first proposed in 2012, a new form of cell death. Autophagy plays a crucial role in cell clearance and maintaining homeostasis. Autophagy is involved in the initial step of ferroptosis under the action of histone elements such as NCOA4, RAB7A, and BECN1. Ferroptosis and autophagy are involved in tumor progression, treatment, and drug resistance in the tumor microenvironment. In this review, we described the mechanisms of ferroptosis, autophagy, and tumor and immunotherapy, respectively, and emphasized the relationship between autophagy-related ferroptosis and tumor.
Collapse
Affiliation(s)
| | | | - Jiale Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Lijuan Du
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yuanyuan Ren
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Fang Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| |
Collapse
|
112
|
Zhang T, Deng W, Deng Y, Liu Y, Xiao S, Luo Y, Xiang W, He Q. Mechanisms of ferroptosis regulating oxidative stress and energy metabolism in myocardial ischemia-reperfusion injury and a novel perspective of natural plant active ingredients for its treatment. Biomed Pharmacother 2023; 165:114706. [PMID: 37400352 DOI: 10.1016/j.biopha.2023.114706] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 07/05/2023] Open
Abstract
Acute myocardial infarction remains the leading cause of death in humans. Timely restoration of blood perfusion to ischemic myocardium remains the most effective strategy in the treatment of acute myocardial infarction, which can significantly reduce morbidity and mortality. However, after restoration of blood flow and reperfusion, myocardial injury will aggravate and induce apoptosis of cardiomyocytes, a process called myocardial ischemia-reperfusion injury. Studies have shown that the loss and death of cardiomyocytes caused by oxidative stress, iron load, increased lipid peroxidation, inflammation and mitochondrial dysfunction, etc., are involved in myocardial ischemia-reperfusion injury. In recent years, with the in-depth research on the pathology of myocardial ischemia-reperfusion injury, people have gradually realized that there is a new form of cell death in the pathological process of myocardial ischemia-reperfusion injury, namely ferroptosis. A number of studies have found that in the myocardial tissue of patients with acute myocardial infarction, there are pathological changes closely related to ferroptosis, such as iron metabolism disorder, lipid peroxidation, and increased reactive oxygen species free radicals. Natural plant products such as resveratrol, baicalin, cyanidin-3-O-glucoside, naringenin, and astragaloside IV can also exert therapeutic effects by correcting the imbalance of these ferroptosis-related factors and expression levels. Combining with our previous studies, this review summarizes the regulatory mechanism of natural plant products intervening ferroptosis in myocardial ischemia-reperfusion injury in recent years, in order to provide reference information for the development of targeted ferroptosis inhibitor drugs for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Tianqing Zhang
- Department of Cardiology, The First People's Hospital of Changde City, Changde 415003, Hunan, China
| | - Wenxu Deng
- The Central Hospital of Hengyang, Hengyang, Hunan 421001, China
| | - Ying Deng
- People's Hospital of Ningxiang City, Ningxiang, Hunan, China
| | - Yao Liu
- The Second Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medcial School, University of South China, Hunan 421001, China.
| | - Sijie Xiao
- Department of Ultrasound, The First People's Hospital of Changde City, Changde 415003, China
| | - Yanfang Luo
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wang Xiang
- Department of Immunology and Rheumatology, The First People's Hospital of Changde City, Changde 415003, China
| | - Qi He
- People's Hospital of Ningxiang City, Ningxiang, Hunan, China
| |
Collapse
|
113
|
Zeng F, Nijiati S, Tang L, Ye J, Zhou Z, Chen X. Ferroptosis Detection: From Approaches to Applications. Angew Chem Int Ed Engl 2023; 62:e202300379. [PMID: 36828775 DOI: 10.1002/anie.202300379] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 02/26/2023]
Abstract
Understanding the intricate molecular machinery that governs ferroptosis and leveraging this accumulating knowledge could facilitate disease prevention, diagnosis, treatment, and prognosis. Emerging approaches for the in situ detection of the major regulators and biological events across cellular, tissue, and in living subjects provide a multiscale perspective for studying ferroptosis. Furthermore, advanced applications that integrate ferroptosis detection and the latest technologies hold tremendous promise in ferroptosis research. In this review, we first briefly summarize the mechanisms and key regulators underlying ferroptosis. Ferroptosis detection approaches are then presented to delineate their design, mechanisms of action, and applications. Special interest is placed on advanced ferroptosis applications that integrate multifunctional platforms. Finally, we discuss the prospects and challenges of ferroptosis detection approaches and applications, with the aim of providing a roadmap for the theranostic development of a broad range of ferroptosis-related diseases.
Collapse
Affiliation(s)
- Fantian Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Sureya Nijiati
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Longguang Tang
- Affiliated Gaozhou People's Hospital, Guangdong Medical University, Guangdong, 524023, China
| | - Jinmin Ye
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zijian Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
114
|
Li Y, He Y, Cheng W, Zhou Z, Ni Z, Yu C. Double-edged roles of ferroptosis in endometriosis and endometriosis-related infertility. Cell Death Discov 2023; 9:306. [PMID: 37607902 PMCID: PMC10444804 DOI: 10.1038/s41420-023-01606-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/31/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023] Open
Abstract
Endometriosis is strongly associated with infertility. Several mechanisms have been reported in an attempt to elucidate the pathophysiological effects that lead to reduced fertility in women with endometriosis. However, the mechanisms by which endometriosis affects fertility have not been fully elucidated. Ferroptosis is a novel form of nonapoptotic cell death that is characterized by iron-dependent lipid peroxidation membrane damage. In past reports, elevated iron levels in ectopic lesions, peritoneal fluid and follicular fluid have been reported in patients with endometriosis. The high-iron environment is closely associated with ferroptosis, which appears to exhibit a double-edged effect on endometriosis. Ferroptosis can cause damage to ovarian granulosa cells, oocytes, and embryos, leading to endometriosis-related infertility. This article summarizes the main pathways and regulatory mechanisms of ferroptosis and explores the possible mechanisms of the formation of an iron-overloaded environment in endometriotic ectopic lesions, peritoneal fluid and follicular fluid. Finally, we reviewed recent studies on the main and potential mechanisms of ferroptosis in endometriosis and endometriosis-related infertility.
Collapse
Affiliation(s)
- Yangshuo Li
- Department of Gynecology of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, 200433, Shanghai, China
| | - Yalun He
- Department of Gynecology of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, 200433, Shanghai, China
| | - Wen Cheng
- Department of Gynecology of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, 200433, Shanghai, China
| | - Zhihao Zhou
- Department of Gynecology of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, 200433, Shanghai, China
| | - Zhexin Ni
- Department of Gynecology of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, 200433, Shanghai, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China.
| | - Chaoqin Yu
- Department of Gynecology of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, 200433, Shanghai, China.
| |
Collapse
|
115
|
Wang X, Tan X, Zhang J, Wu J, Shi H. The emerging roles of MAPK-AMPK in ferroptosis regulatory network. Cell Commun Signal 2023; 21:200. [PMID: 37580745 PMCID: PMC10424420 DOI: 10.1186/s12964-023-01170-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/20/2023] [Indexed: 08/16/2023] Open
Abstract
Ferroptosis, a newform of programmed cell death, driven by peroxidative damages of polyunsaturated-fatty-acid-containing phospholipids in cellular membranes and is extremely dependent on iron ions, which is differs characteristics from traditional cell death has attracted greater attention. Based on the curiosity of this new form of regulated cell death, there has a tremendous progress in the field of mechanistic understanding of ferroptosis recent years. Ferroptosis is closely associated with the development of many diseases and involved in many diseases related signaling pathways. Not only a variety of oncoproteins and tumor suppressors can regulate ferroptosis, but multiple oncogenic signaling pathways can also have a regulatory effect on ferroptosis. Ferroptosis results in the accumulation of large amounts of lipid peroxides thus involving the onset of oxidative stress and energy stress responses. The MAPK pathway plays a critical role in oxidative stress and AMPK acts as a sensor of cellular energy and is involved in the regulation of the energy stress response. Moreover, activation of AMPK can induce the occurrence of autophagy-dependent ferroptosis and p53-activated ferroptosis. In recent years, there have been new advances in the study of molecular mechanisms related to the regulation of ferroptosis by both pathways. In this review, we will summarize the molecular mechanisms by which the MAPK-AMPK signaling pathway regulates ferroptosis. Meanwhile, we sorted out the mysterious relationship between MAPK and AMPK, described the crosstalk among ferroptosis and MAPK-AMPK signaling pathways, and summarized the relevant ferroptosis inducers targeting this regulatory network. This will provide a new field for future research on ferroptosis mechanisms and provide a new vision for cancer treatment strategies. Video Abstract.
Collapse
Affiliation(s)
- Xinyue Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Xiao Tan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Jinping Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Jiaping Wu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Hongjuan Shi
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| |
Collapse
|
116
|
Sheng SY, Li JM, Hu XY, Wang Y. Regulated cell death pathways in cardiomyopathy. Acta Pharmacol Sin 2023; 44:1521-1535. [PMID: 36914852 PMCID: PMC10374591 DOI: 10.1038/s41401-023-01068-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
Heart disease is a worldwide health menace. Both intractable primary and secondary cardiomyopathies contribute to malignant cardiac dysfunction and mortality. One of the key cellular processes associated with cardiomyopathy is cardiomyocyte death. Cardiomyocytes are terminally differentiated cells with very limited regenerative capacity. Various insults can lead to irreversible damage of cardiomyocytes, contributing to progression of cardiac dysfunction. Accumulating evidence indicates that majority of cardiomyocyte death is executed by regulating molecular pathways, including apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Importantly, these forms of regulated cell death (RCD) are cardinal features in the pathogenesis of various cardiomyopathies, including dilated cardiomyopathy, diabetic cardiomyopathy, sepsis-induced cardiomyopathy, and drug-induced cardiomyopathy. The relevance between abnormity of RCD with adverse outcome of cardiomyopathy has been unequivocally evident. Therefore, there is an urgent need to uncover the molecular and cellular mechanisms for RCD in order to better understand the pathogenesis of cardiomyopathies. In this review, we summarize the latest progress from studies on RCD pathways in cardiomyocytes in context of the pathogenesis of cardiomyopathies, with particular emphasis on apoptosis, necroptosis, ferroptosis, autophagy, and pyroptosis. We also elaborate the crosstalk among various forms of RCD in pathologically stressed myocardium and the prospects of therapeutic applications targeted to various cell death pathways.
Collapse
Affiliation(s)
- Shu-Yuan Sheng
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Jia-Min Li
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Xin-Yang Hu
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Yibin Wang
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China.
- Signature Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School and National Heart Center of Singapore, Singapore, Singapore.
| |
Collapse
|
117
|
Zheng X, Jin X, Ye F, Liu X, Yu B, Li Z, Zhao T, Chen W, Liu X, Di C, Li Q. Ferroptosis: a novel regulated cell death participating in cellular stress response, radiotherapy, and immunotherapy. Exp Hematol Oncol 2023; 12:65. [PMID: 37501213 PMCID: PMC10375783 DOI: 10.1186/s40164-023-00427-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Ferroptosis is a regulated cell death mode triggered by iron-dependent toxic membrane lipid peroxidation. As a novel cell death modality that is morphologically and mechanistically different from other forms of cell death, such as apoptosis and necrosis, ferroptosis has attracted extensive attention due to its association with various diseases. Evidence on ferroptosis as a potential therapeutic strategy has accumulated with the rapid growth of research on targeting ferroptosis for tumor suppression in recent years. METHODS We summarize the currently known characteristics and major regulatory mechanisms of ferroptosis and present the role of ferroptosis in cellular stress responses, including ER stress and autophagy. Furthermore, we elucidate the potential applications of ferroptosis in radiotherapy and immunotherapy, which will be beneficial in exploring new strategies for clinical tumor treatment. RESULT AND CONCLUSION Based on specific biomarkers and precise patient-specific assessment, targeting ferroptosis has great potential to be translated into practical new approaches for clinical cancer therapy, significantly contributing to the prevention, diagnosis, prognosis, and treatment of cancer.
Collapse
Affiliation(s)
- Xiaogang Zheng
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaodong Jin
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei Ye
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiongxiong Liu
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Boyi Yu
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zheng Li
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ting Zhao
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weiqiang Chen
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinguo Liu
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cuixia Di
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiang Li
- Department of Medical Physics, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
118
|
Yan X, Xie Y, Liu H, Huang M, Yang Z, An D, Jiang G. Iron accumulation and lipid peroxidation: implication of ferroptosis in diabetic cardiomyopathy. Diabetol Metab Syndr 2023; 15:161. [PMID: 37468902 DOI: 10.1186/s13098-023-01135-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023] Open
Abstract
Diabetic cardiomyopathy (DC) is a serious heart disease caused by diabetes. It is unrelated to hypertension and coronary artery disease and can lead to heart insufficiency, heart failure and even death. Currently, the pathogenesis of DC is unclear, and clinical intervention is mainly symptomatic therapy and lacks effective intervention objectives. Iron overdose mediated cell death, also known as ferroptosis, is widely present in the physiological and pathological processes of diabetes and DC. Iron is a key trace element in the human body, regulating the metabolism of glucose and lipids, oxidative stress and inflammation, and other biological processes. Excessive iron accumulation can lead to the imbalance of the antioxidant system in DC and activate and aggravate pathological processes such as excessive autophagy and mitochondrial dysfunction, resulting in a chain reaction and accelerating myocardial and microvascular damage. In-depth understanding of the regulating mechanisms of iron metabolism and ferroptosis in cardiovascular vessels can help improve DC management. Therefore, in this review, we summarize the relationship between ferroptosis and the pathogenesis of DC, as well as potential intervention targets, and discuss and analyze the limitations and future development prospects of these targets.
Collapse
Affiliation(s)
- Xuehua Yan
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Xinjiang, China
| | - Yang Xie
- Affiliated Hospital of Traditional Chinese Medicine of Xinjiang Medical University, Xinjiang, China
| | - Hongbing Liu
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
| | - Meng Huang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
| | - Zhen Yang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
| | - Dongqing An
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China.
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Xinjiang, China.
- Affiliated Hospital of Traditional Chinese Medicine of Xinjiang Medical University, Xinjiang, China.
| | - Guangjian Jiang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China.
| |
Collapse
|
119
|
Zhang XR, Luo YT, Zhu FY, Ye L, Ni SY, Liu YC, Shen Y. Novel target for treatment of colorectal cancer: Metabolism and regulatory mechanisms of ferroptosis. Shijie Huaren Xiaohua Zazhi 2023; 31:528-536. [DOI: 10.11569/wcjd.v31.i13.528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/20/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023] Open
Abstract
Ferroptosis is a new type of programmed cell death charac-terized by intracellular iron overload and lipid peroxidation accumulation. In the past 10 years, research has demonstrated that intervention of ferroptosis can effectively prevent or treat cancer diseases, including colorectal cancer. However, the role of ferroptosis in different types of colorectal cancer is not the same. This article summarizes the three major pathways involved in the occurrence and development of ferroptosis, namely, iron metabolism, lipid metabolism, and amino acid metabolism, and discusses the specific mechanisms and clinical applications of ferroptosis targeted therapy in colorectal cancer of various consensus molecular subtypes. Existing studies have shown that ferroptosis targeted therapy has broad application prospects in optimizing the comprehensive treatment of colorectal cancer. This article will provide an important framework for studying the mechanism of ferroptosis in tumor prevention and treatment and have guiding significance in exploring the application of ferroptosis targeted therapy in the treatment of different types of colorectal cancer.
Collapse
Affiliation(s)
- Xin-Rui Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Yi-Ting Luo
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Fang-Yuan Zhu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Lu Ye
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Si-Yi Ni
- Department of Gastroente-rology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Ying-Chao Liu
- Department of Gastroente-rology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Yan Shen
- Department of Gastroente-rology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
120
|
Lai W, Chen J, Wang T, Liu Q. Crosstalk between ferroptosis and steroid hormone signaling in gynecologic cancers. Front Mol Biosci 2023; 10:1223493. [PMID: 37469703 PMCID: PMC10352791 DOI: 10.3389/fmolb.2023.1223493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/22/2023] [Indexed: 07/21/2023] Open
Abstract
Ferroptosis is a novel types of regulated cell death and is widely studied in cancers and many other diseases in recent years. It is characterized by iron accumulation and intense lipid peroxidation that ultimately inducing oxidative damage. So far, signaling pathways related to ferroptosis are involved in all aspects of determining cell fate, including oxidative phosphorylation, metal-ion transport, energy metabolism and cholesterol synthesis progress, et al. Recently, accumulated studies have demonstrated that ferroptosis is associated with gynecological oncology related to steroid hormone signaling. This review trends to summarize the mechanisms and applications of ferroptosis in cancers related to estrogen and progesterone, which is expected to provide a theoretical basis for the prevention and treatment of gynecologic cancers.
Collapse
Affiliation(s)
- Wen Lai
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Tianming Wang
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Qiaoling Liu
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
121
|
Posadino AM, Giordo R, Pintus G, Mohammed SA, Orhan IE, Fokou PVT, Sharopov F, Adetunji CO, Gulsunoglu-Konuskan Z, Ydyrys A, Armstrong L, Sytar O, Martorell M, Razis AFA, Modu B, Calina D, Habtemariam S, Sharifi-Rad J, Cho WC. Medicinal and mechanistic overview of artemisinin in the treatment of human diseases. Biomed Pharmacother 2023; 163:114866. [PMID: 37182516 DOI: 10.1016/j.biopha.2023.114866] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023] Open
Abstract
Artemisinin (ART) is a bioactive compound isolated from the plant Artemisia annua and has been traditionally used to treat conditions such as malaria, cancer, viral infections, bacterial infections, and some cardiovascular diseases, especially in Asia, North America, Europe and other parts of the world. This comprehensive review aims to update the biomedical potential of ART and its derivatives for treating human diseases highlighting its pharmacokinetic and pharmacological properties based on the results of experimental pharmacological studies in vitro and in vivo. Cellular and molecular mechanisms of action, tested doses and toxic effects of artemisinin were also described. The analysis of data based on an up-to-date literature search showed that ART and its derivatives display anticancer effects along with a wide range of pharmacological activities such as antibacterial, antiviral, antimalarial, antioxidant and cardioprotective effects. These compounds have great potential for discovering new drugs used as adjunctive therapies in cancer and various other diseases. Detailed translational and experimental studies are however needed to fully understand the pharmacological effects of these compounds.
Collapse
Affiliation(s)
- Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy
| | - Roberta Giordo
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy; Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah 27272, United Arab Emirates
| | - Soheb Anwar Mohammed
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, University of Pittsburgh, PA 15213, USA
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330 Ankara, Turkey; Turkish Academy of Sciences (TÜBA), Vedat Dalokay Cad., No. 112, 06670 Ankara, Turkey
| | | | - Farukh Sharopov
- V.I. Nikitin Chemistry Institute of the National Academy of Sciences of Tajikistan, Ayni 299/2, 734063 Dushanbe, Tajikistan
| | - Charles Oluwaseun Adetunji
- Applied Microbiology, Biotechnology and Nanotechnology Laboratory, Department of Microbiology, Edo State University Uzairue, Iyamho, PMB 04 Auchi, Edo State, Nigeria
| | - Zehra Gulsunoglu-Konuskan
- Faculty of Health Science, Nutrition and Dietetics Department, Istanbul Aydin University, Istanbul 34295, Turkey
| | - Alibek Ydyrys
- Biomedical Research Centre, Al-Farabi Kazakh National University, Al-Farabi ave. 71, 050040 Almaty, Kazakhstan
| | - Lorene Armstrong
- State University of Ponta Grossa, Departament of Pharmaceutical Sciences, 84030900 Ponta Grossa, Paraná, Brazil; Federal University of Paraná, Department of Pharmacy, 80210170 Curitiba, Paraná, Brazil
| | - Oksana Sytar
- Institute of Plant and Environmental Sciences, Slovak Agricultural University in Nitra, 94976 Nitra, Slovakia
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepción, 4070386 Concepción, Chile; Universidad de Concepción, Unidad de Desarrollo Tecnológico, UDT, 4070386 Concepción, Chile.
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Babagana Modu
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Department of Biochemistry, Faculty of Science, University of Maiduguri, 1069 Maiduguri, Borno State, Nigeria
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| | | | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
122
|
Wu H, Liu Q, Shan X, Gao W, Chen Q. ATM orchestrates ferritinophagy and ferroptosis by phosphorylating NCOA4. Autophagy 2023; 19:2062-2077. [PMID: 36752571 PMCID: PMC10283418 DOI: 10.1080/15548627.2023.2170960] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 02/09/2023] Open
Abstract
Ferroptosis is a newly characterized form of programmed cell death, which is driven by the lethal accumulation of lipid peroxides catalyzed by the intracellular bioactive iron. Targeted induction of ferroptotic cell death holds great promise for therapeutic design against other therapy-resistant cancers. To date, multiple post-translational modifications have been elucidated to impinge on the ferroptotic sensitivity. Here we report that the Ser/Thr protein kinase ATM, the major sensor of DNA double-strand break damage, is indispensable for ferroptosis execution. Pharmacological inhibition or genetic ablation of ATM significantly antagonizes ferroptosis. Besides, ATM ablation-induced ferroptotic resistance is largely independent of its downstream target TRP53, as cells defective in both Trp53 and Atm are still more insensitive to ferroptotic inducers than the trp53 single knockout cells. Mechanistically, ATM dominates the intracellular labile free iron by phosphorylating NCOA4, facilitating NCOA4-ferritin interaction and therefore sustaining ferritinophagy, a selective type of macroautophagy/autophagy specifically degrading ferritin for iron recycling. Our results thus uncover a novel regulatory circuit of ferroptosis comprising ATM-NCOA4 in orchestrating ferritinophagy and iron bioavailability.Abbreviations: AMPK: AMP-activated protein kinase; ATM: ataxia telangiectasia mutated; BSO: buthionine sulphoximine; CDKN1A: cyclin-dependent kinase inhibitor 1A (P21); CQ: chloroquine; DFO: deferoxamine; DFP: deferiprone; Fer: ferrostatin-1; FTH1: ferritin heavy polypeptide 1; GPX4: glutathione peroxidase 4; GSH: glutathione; MEF: mouse embryonic fibroblast; NCOA4: nuclear receptor coactivator 4; PFTα: pifithrin-α; PTGS2: prostaglandin-endoperoxide synthase 2; Slc7a11: solute carrier family 7 member 11; Sul: sulfasalazine; TFRC: transferrin receptor; TRP53: transformation related protein 53.
Collapse
Affiliation(s)
- Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- Interdisciplinary Sciences Research Institute, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qian Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- Interdisciplinary Sciences Research Institute, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xinyi Shan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- Interdisciplinary Sciences Research Institute, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Weihua Gao
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- Interdisciplinary Sciences Research Institute, Huazhong Agricultural University, Wuhan, Hubei, China
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Quan Chen
- State key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
123
|
Liu J, Hu Z, Ma Q, Wang S, Liu D. Ferritin-dependent cellular autophagy pathway promotes ferroptosis in beef during cold storage. Food Chem 2023; 412:135550. [PMID: 36706507 DOI: 10.1016/j.foodchem.2023.135550] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
Ferroptosis plays a pivotal role in regulating various physiological processes and quality of post-mortem muscle. However, the molecular mechanisms underlying ferroptosis remain unclear. The study investigated how ferroptosis was induced in beef during cold storage. Results showed that the expression of autophagy-related genes, LC3, ATG5, ATG7, and NCOA4 in beef during cold storage promoted the degradation of ferritin heavy chains. Ferritin evoked ferroptosis by releasing free iron, inducing reactive oxygen species (ROS) accumulation and inhibiting the glutathione (GSH)-glutathione peroxidase 4 (GPX4) pathway. Furthermore, treatment of myoblasts with GSK 2656157 (autophagy inhibitor) showed that ferritin degradation was lower in the GSK 2656157-treated myoblasts than in the control, while GSH content and GPX4 activity were higher than the control (P < 0.05), and the contents of free iron, ROS and malondialdehyde, and apoptosis were lower than the control (P < 0.05). These results suggest that ferroptosis is induced by degradation of ferritin via the autophagic pathway.
Collapse
Affiliation(s)
- Jun Liu
- School of Agriculture, Ningxia University, 750021 Yinchuan, China
| | - Ziying Hu
- School of Food & Wine, Ningxia University, 750021 Yinchuan, China
| | - Qin Ma
- School of Food & Wine, Ningxia University, 750021 Yinchuan, China
| | - Shuzhe Wang
- School of Agriculture, Ningxia University, 750021 Yinchuan, China; Ningxia Key Laboratory of Ruminant Molecular and Cellular Breeding, Ningxia University, 750021 Yinchuan, China
| | - Dunhua Liu
- School of Agriculture, Ningxia University, 750021 Yinchuan, China; School of Food & Wine, Ningxia University, 750021 Yinchuan, China.
| |
Collapse
|
124
|
Tian C, Zheng M, Lan X, Liu L, Ye Z, Li C. Silencing LCN2 enhances RSL3-induced ferroptosis in T cell acute lymphoblastic leukemia. Gene 2023:147597. [PMID: 37390872 DOI: 10.1016/j.gene.2023.147597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND T-cell acute lymphoblastic leukemia (T-ALL) is a life-threatening malignancy and therapeutic toxicity remains a huge challenge for survival rates. A novel iron-dependent form of cell death, ferroptosis, shows potentials in cancer therapy. This study aimed to identify ferroptosis-associated hub genes within a proteinprotein interaction (PPI) network. METHODS We screened differential expressed genes (DEGs) in GSE46170 dataset and obtained ferroptosis-related genes from FerrDb database. Through overlapping between DEGs and ferroptosis-related genes, ferroptosis-associated DEGs were identified for further PPI network construction. Molecular complex detection (MCODE) algorithm in Cytoscape was employed to determine tightly connected protein clusters. Chord diagram of Gene Ontology (GO) was generated to reveal the potential biological process of hub genes. Through transfection with siRNA of lipocalin 2 (LCN2) into TALL cells, the regulatory role of LCN2 in ferroptosis was investigated. RESULTS Venn diagram identified a total of 37 ferroptosis-associated DEGs between GSE46170 and ferroptosis-associated genes, which were mainly enriched in ferroptosis and necroptosis. Based on PPI network analysis, 5 hub genes (LCN2, LTF, HP, SLC40A1 and TFRC) were found. These hub genes were involved in iron ion transport and could distinguish T-ALL from normal individuals. Further experimental studies demonstrated that LCN2 was highly expressed in T-ALL, while silencing LCN2 promoted RSL3-induced ferroptotic cell death in T-ALL cells. CONCLUSION This study identified novel ferroptosis-associated hub genes, which shed new insights into the underlying mechanism of ferroptosis in T-ALL and also provide promising therapeutic targets for T-ALL.
Collapse
Affiliation(s)
- Chuan Tian
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, No. 57, South Renmin Avenue, Zhanjiang City, Guangdong Province, 524001, China
| | - Min Zheng
- Department of Obstetrics, Affiliated Hospital of Guangdong Medical University, No. 57, South Renmin Avenue, Zhanjiang City, Guangdong Province, 524001, China
| | - Xiang Lan
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, No. 57, South Renmin Avenue, Zhanjiang City, Guangdong Province, 524001, China
| | - Lili Liu
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, No. 57, South Renmin Avenue, Zhanjiang City, Guangdong Province, 524001, China
| | - Zhonglv Ye
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, No. 57, South Renmin Avenue, Zhanjiang City, Guangdong Province, 524001, China
| | - Chengyan Li
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, No. 57, South Renmin Avenue, Zhanjiang City, Guangdong Province, 524001, China.
| |
Collapse
|
125
|
Tang X, Wang Y, Zhu Y, Guo Y, Liu B. Basic mechanisms and novel potential therapeutic targets for ferroptosis in acute myeloid leukemia. Ann Hematol 2023:10.1007/s00277-023-05293-4. [PMID: 37314462 DOI: 10.1007/s00277-023-05293-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/22/2023] [Indexed: 06/15/2023]
Abstract
Ferroptosis is a form of cell death that is regulated by iron and characterized by the buildup of lipid peroxides (LPO) and subsequent rupture of the cell membrane. The molecular mechanisms of ferroptosis involve metabolic pathways related to iron, lipids, and amino acids, which contribute to the production of lipid reactive oxygen species (ROS). In recent years, there has been increasing attention on the occurrence of ferroptosis in various diseases. Ferroptosis has been found to play a crucial role in cardiovascular diseases, digestive diseases, respiratory and immunological diseases, and particularly in malignancies. However, there is still a lack of studies on ferroptosis in acute myeloid leukemia (AML). This paper provides a comprehensive review of the mechanism of ferroptosis and its regulatory molecules and therapeutic agents in AML. It also evaluates the relationship between ferroptosis-related genes (FRGs), non-coding RNAs (ncRNAs), and prognosis to develop prognostic molecular models in AML. The study also explores the association between ferroptosis and immune infiltration in AML, to identify novel potential target regimens for AML.
Collapse
Affiliation(s)
- Xiao Tang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730099, China
| | - Yin Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730099, China
| | - Yu Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730099, China
| | - Yuancheng Guo
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730099, China
| | - Bei Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730099, China.
- Department of Hematology, The First Affiliated Hospital, Lanzhou University, Lanzhou, 730099, China.
| |
Collapse
|
126
|
Cheng X, Zhang J, Xiao Y, Wang Z, He J, Ke M, Liu S, Wang Q, Zhang L. Mitochondrial Regulation of Ferroptosis in Cancer Therapy. Int J Mol Sci 2023; 24:10037. [PMID: 37373183 DOI: 10.3390/ijms241210037] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Ferroptosis, characterized by glutamate overload, glutathione depletion, and cysteine/cystine deprivation during iron- and oxidative-damage-dependent cell death, is a particular mode of regulated cell death. It is expected to effectively treat cancer through its tumor-suppressor function, as mitochondria are the intracellular energy factory and a binding site of reactive oxygen species production, closely related to ferroptosis. This review summarizes relevant research on the mechanisms of ferroptosis, highlights mitochondria's role in it, and collects and classifies the inducers of ferroptosis. A deeper understanding of the relationship between ferroptosis and mitochondrial function may provide new strategies for tumor treatment and drug development based on ferroptosis.
Collapse
Affiliation(s)
- Xiaoxia Cheng
- School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Jiale Zhang
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Yichen Xiao
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Zhihang Wang
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Jin He
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Mengquan Ke
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Sijie Liu
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Qun Wang
- School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Lei Zhang
- School of Basic Medical Science, Henan University, Kaifeng 475004, China
| |
Collapse
|
127
|
Jing Q, Yuan C, Zhou C, Jin W, Wang A, Wu Y, Shang W, Zhang G, Ke X, Du J, Li Y, Shao F. Comprehensive analysis identifies CLEC1B as a potential prognostic biomarker in hepatocellular carcinoma. Cancer Cell Int 2023; 23:113. [PMID: 37308868 PMCID: PMC10262401 DOI: 10.1186/s12935-023-02939-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/06/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND C-type lectin domain family 1 member B (CLEC1B, encoding the CLEC-2 protein), a member of the C-type lectin superfamily, is a type II transmembrane receptor involved in platelet activation, angiogenesis, and immune and inflammatory responses. However, data regarding its function and clinical prognostic value in hepatocellular carcinoma (HCC) remain scarce. METHODS The expression of CLEC1B was explored using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. RT-qPCR, western blot, and immunohistochemistry assays were employed to validate the downregulation of CLEC1B. Univariate Cox regression and survival analyses were used to evaluate the prognostic value of CLEC1B. Gene Set Enrichment Analysis (GSEA) was conducted to investigate the potential association between cancer hallmarks and CLEC1B expression. The TISIDB database was applied to search for the correlation between immune cell infiltration levels and CLEC1B expression. The association between CLEC1B and immunomodulators was conducted by Spearman correlation analysis based on the Sangerbox platform. Annexin V-FITC/PI apoptosis kit was used for the detection of cell apoptosis. RESULTS The expression of CLEC1B was low in various tumors and exhibited a promising clinical prognostic value for HCC patients. The expression level of CLEC1B was tightly associated with the infiltration of various immune cells in the HCC tumor microenvironment (TME) and positively correlated with a bulk of immunomodulators. In addition, CLEC1B and its related genes or interacting proteins are implicated in multiple immune-related processes and signaling pathways. Moreover, overexpression of CLEC1B significantly influenced the treatment effects of sorafenib on HCC cells. CONCLUSIONS Our results reveal that CLEC1B could serve as a potential prognostic biomarker and may be a novel immunoregulator for HCC. However, its function in immune regulation should be further explored.
Collapse
Grants
- 2021KY077, 2022KY503, 2022KY046, 2022KY074, 2022KY290 Medical and Health Science and Technology Project of Zhejiang Province
- 2021KY077, 2022KY503, 2022KY046, 2022KY074, 2022KY290 Medical and Health Science and Technology Project of Zhejiang Province
- 2021KY077, 2022KY503, 2022KY046, 2022KY074, 2022KY290 Medical and Health Science and Technology Project of Zhejiang Province
- 2020ZA098, 2021ZB245 Traditional Chinese Medicine Science and Technology Project of Zhejiang Province
- 2020ZA098, 2021ZB245 Traditional Chinese Medicine Science and Technology Project of Zhejiang Province
- LGF21H010008, LGF20H080005, LBY23H080004, LGF22H080008 Zhejiang Provincial Natural Science Foundation of China
- LGF21H010008, LGF20H080005, LBY23H080004, LGF22H080008 Zhejiang Provincial Natural Science Foundation of China
- LGF21H010008, LGF20H080005, LBY23H080004, LGF22H080008 Zhejiang Provincial Natural Science Foundation of China
Collapse
Affiliation(s)
- Qiangan Jing
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Chen Yuan
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chaoting Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Weidong Jin
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Aiwei Wang
- Department of Hematology, The first people's Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang, China
| | - Yanfang Wu
- Department of Hematology, The first people's Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang, China
| | - Wenzhong Shang
- Department of Hematology, The first people's Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang, China
| | - Guibing Zhang
- Department of Hematology, The first people's Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang, China
| | - Xia Ke
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Fangchun Shao
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
128
|
Saulle E, Spinello I, Quaranta MT, Labbaye C. Advances in Understanding the Links between Metabolism and Autophagy in Acute Myeloid Leukemia: From Biology to Therapeutic Targeting. Cells 2023; 12:1553. [PMID: 37296673 PMCID: PMC10252746 DOI: 10.3390/cells12111553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Autophagy is a highly conserved cellular degradation process that regulates cellular metabolism and homeostasis under normal and pathophysiological conditions. Autophagy and metabolism are linked in the hematopoietic system, playing a fundamental role in the self-renewal, survival, and differentiation of hematopoietic stem and progenitor cells, and in cell death, particularly affecting the cellular fate of the hematopoietic stem cell pool. In leukemia, autophagy sustains leukemic cell growth, contributes to survival of leukemic stem cells and chemotherapy resistance. The high frequency of disease relapse caused by relapse-initiating leukemic cells resistant to therapy occurs in acute myeloid leukemia (AML), and depends on the AML subtypes and treatments used. Targeting autophagy may represent a promising strategy to overcome therapeutic resistance in AML, for which prognosis remains poor. In this review, we illustrate the role of autophagy and the impact of its deregulation on the metabolism of normal and leukemic hematopoietic cells. We report updates on the contribution of autophagy to AML development and relapse, and the latest evidence indicating autophagy-related genes as potential prognostic predictors and drivers of AML. We review the recent advances in autophagy manipulation, combined with various anti-leukemia therapies, for an effective autophagy-targeted therapy for AML.
Collapse
Affiliation(s)
- Ernestina Saulle
- Correspondence: (E.S.); (C.L.); Tel.: +39-0649902422 (E.S.); +39-0649902418 (C.L.)
| | | | | | - Catherine Labbaye
- Correspondence: (E.S.); (C.L.); Tel.: +39-0649902422 (E.S.); +39-0649902418 (C.L.)
| |
Collapse
|
129
|
Xie Y, Lei X, Zhao G, Guo R, Cui N. mTOR in programmed cell death and its therapeutic implications. Cytokine Growth Factor Rev 2023; 71-72:66-81. [PMID: 37380596 DOI: 10.1016/j.cytogfr.2023.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Mechanistic target of rapamycin (mTOR), a highly conserved serine/threonine kinase, is involved in cellular metabolism, protein synthesis, and cell death. Programmed cell death (PCD) assists in eliminating aging, damaged, or neoplastic cells, and is indispensable for sustaining normal growth, fighting pathogenic microorganisms, and maintaining body homeostasis. mTOR has crucial functions in the intricate signaling pathway network of multiple forms of PCD. mTOR can inhibit autophagy, which is part of PCD regulation. Cell survival is affected by mTOR through autophagy to control reactive oxygen species production and the degradation of pertinent proteins. Additionally, mTOR can regulate PCD in an autophagy-independent manner by affecting the expression levels of related genes and phosphorylating proteins. Therefore, mTOR acts through both autophagy-dependent and -independent pathways to regulate PCD. It is conceivable that mTOR exerts bidirectional regulation of PCD, such as ferroptosis, according to the complexity of signaling pathway networks, but the underlying mechanisms have not been fully explained. This review summarizes the recent advances in understanding mTOR-mediated regulatory mechanisms in PCD. Rigorous investigations into PCD-related signaling pathways have provided prospective therapeutic targets that may be clinically beneficial for treating various diseases.
Collapse
Affiliation(s)
- Yawen Xie
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xianli Lei
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoyu Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ran Guo
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
130
|
Lee J, Roh JL. Targeting Iron-Sulfur Clusters in Cancer: Opportunities and Challenges for Ferroptosis-Based Therapy. Cancers (Basel) 2023; 15:2694. [PMID: 37345031 DOI: 10.3390/cancers15102694] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Iron dysregulation is a hallmark of cancer, characterized by an overexpression of genes involved in iron metabolism and iron-sulfur cluster (ISC) biogenesis. Dysregulated iron homeostasis increases intracellular labile iron, which may lead to the formation of excess cytotoxic radicals and make it vulnerable to various types of regulated cell death, including ferroptosis. The inhibition of ISC synthesis triggers the iron starvation response, increasing lipid peroxidation and ferroptosis in cancer cells treated with oxidative stress-inducing agents. Various methods, such as redox operations, iron chelation, and iron replacement with redox-inert metals, can destabilize or limit ISC formation and function, providing potential therapeutic strategies for cancer treatment. Targeting ISCs to induce ferroptosis represents a promising approach in cancer therapy. This review summarizes the state-of-the-art overview of iron metabolism and ferroptosis in cancer cells, the role of ISC modulation in ferroptosis, and the potential of targeting ISCs for ferroptosis induction in cancer therapy. Further research is necessary to develop and validate these strategies in clinical trials for various cancers, which may ultimately lead to the development of novel and effective treatments for cancer patients.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam 13488, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam 13488, Republic of Korea
| |
Collapse
|
131
|
Zhou L, Jia X, Shang Y, Sun Y, Liu Z, Liu J, Jiang W, Deng S, Yao Q, Chen J, Li H. PRMT1 inhibition promotes ferroptosis sensitivity via ACSL1 upregulation in acute myeloid leukemia. Mol Carcinog 2023. [PMID: 37144835 DOI: 10.1002/mc.23550] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 03/24/2023] [Accepted: 04/13/2023] [Indexed: 05/06/2023]
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy with an alarming mortality rate. The development of novel therapeutic targets or drugs for AML is urgently needed. Ferroptosis is a form of regulated cell death driven by iron-dependent lipid peroxidation. Recently, ferroptosis has emerged as a novel method for targeting cancer, including AML. Epigenetic dysregulation is a hallmark of AML, and a growing body of evidence suggests that ferroptosis is subject to epigenetic regulation. Here, we identified protein arginine methyltransferase 1 (PRMT1) as a ferroptosis regulator in AML. The type I PRMT inhibitor GSK3368715 promoted ferroptosis sensitivity in vitro and in vivo. Moreover, PRMT1-knockout cells exhibited significantly increased sensitivity to ferroptosis, suggesting that PRMT1 is the primary target of GSK3368715 in AML. Mechanistically, both GSK3368715 and PRMT1 knockout upregulated acyl-CoA synthetase long-chain family member 1 (ACSL1), which acts as a ferroptosis promoter by increasing lipid peroxidation. Knockout ACSL1 reduced the ferroptosis sensitivity of AML cells following GSK3368715 treatment. Additionally, the GSK3368715 treatment reduced the abundance of H4R3me2a, the main histone methylation modification mediated by PRMT1, in both genome-wide and ACSL1 promoter regions. Overall, our results demonstrated a previously unknown role of the PRMT1/ACSL1 axis in ferroptosis and suggested the potential value and applications of the combination of PRMT1 inhibitor and ferroptosis inducers in AML treatment.
Collapse
Affiliation(s)
- Lixin Zhou
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaoqing Jia
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yingying Shang
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yanni Sun
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhilong Liu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jifeng Liu
- Department of Anus-Intestines, The People's Hospital of Luzhou, Luzhou, China
| | - Wen Jiang
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Siyuan Deng
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qi Yao
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jieping Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Li
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
132
|
Fujii J, Yamada KI. Defense systems to avoid ferroptosis caused by lipid peroxidation-mediated membrane damage. Free Radic Res 2023; 57:353-372. [PMID: 37551716 DOI: 10.1080/10715762.2023.2244155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
The presence of hydrogen peroxide along with ferrous iron produces hydroxyl radicals that preferably oxidize polyunsaturated fatty acids (PUFA) to alkyl radicals (L•). The reaction of L• with an oxygen molecule produces lipid peroxyl radical (LOO•) that collectively trigger chain reactions, which results in the accumulation of lipid peroxidation products (LOOH). Oxygenase enzymes, such as lipoxygenase, also stimulate the peroxidation of PUFA. The production of phospholipid hydroperoxides (P-LOOH) can result in the destruction of the architecture of cell membranes and ultimate cell death. This iron-dependent regulated cell death is generally referred to as ferroptosis. Radical scavengers, which include tocopherol and nitric oxide (•NO), react with lipid radicals and terminate the chain reaction. When tocopherol reductively detoxifies lipid radicals, the resultant tocopherol radicals are recycled via reduction by coenzyme Q or ascorbate. CoQ radicals are reduced back by the anti-ferroptotic enzyme FSP1. •NO reacts with lipid radicals and produces less reactive nitroso compounds. The resulting P-LOOH is reductively detoxified by the action of glutathione peroxidase 4 (GPX4) or peroxiredoxin 6 (PRDX6). The hydrolytic removal of LOOH from P-LOOH by calcium-independent phospholipase A2 leads the preservation of membrane structure. While the expression of such protective genes or the presence of these anti-oxidant compounds serve to maintain a healthy condition, tumor cells employ them to make themselves resistant to anti-tumor treatments. Thus, these defense mechanisms against ferroptosis are protective in ordinary cells but are also potential targets for cancer treatment.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Ken-Ichi Yamada
- Faculty of Pharmaceutical Sciences, Physical Chemistry for Life Science Laboratory, Kyushu University, Fukuoka, Japan
| |
Collapse
|
133
|
Duan Y, Sun J. Preparation of Iron-Based Sulfides and Their Applications in Biomedical Fields. Biomimetics (Basel) 2023; 8:biomimetics8020177. [PMID: 37218763 DOI: 10.3390/biomimetics8020177] [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: 03/15/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Recently, iron-based sulfides, including iron sulfide minerals and biological iron sulfide clusters, have attracted widespread interest, owing to their excellent biocompatibility and multi-functionality in biomedical applications. As such, controlled synthesized iron sulfide nanomaterials with elaborate designs, enhanced functionality and unique electronic structures show numerous advantages. Furthermore, iron sulfide clusters produced through biological metabolism are thought to possess magnetic properties and play a crucial role in balancing the concentration of iron in cells, thereby affecting ferroptosis processes. The electrons in the Fenton reaction constantly transfer between Fe2+ and Fe3+, participating in the production and reaction process of reactive oxygen species (ROS). This mechanism is considered to confer advantages in various biomedical fields such as the antibacterial field, tumor treatment, biosensing and the treatment of neurodegenerative diseases. Thus, we aim to systematically introduce recent advances in common iron-based sulfides.
Collapse
Affiliation(s)
- Yefan Duan
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210009, China
| | - Jianfei Sun
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210009, China
| |
Collapse
|
134
|
Mynott RL, Habib A, Best OG, Wallington-Gates CT. Ferroptosis in Haematological Malignancies and Associated Therapeutic Nanotechnologies. Int J Mol Sci 2023; 24:ijms24087661. [PMID: 37108836 PMCID: PMC10146166 DOI: 10.3390/ijms24087661] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Haematological malignancies are heterogeneous groups of cancers of the bone marrow, blood or lymph nodes, and while therapeutic advances have greatly improved the lifespan and quality of life of those afflicted, many of these cancers remain incurable. The iron-dependent, lipid oxidation-mediated form of cell death, ferroptosis, has emerged as a promising pathway to induce cancer cell death, particularly in those malignancies that are resistant to traditional apoptosis-inducing therapies. Although promising findings have been published in several solid and haematological malignancies, the major drawbacks of ferroptosis-inducing therapies are efficient drug delivery and toxicities to healthy tissue. The development of tumour-targeting and precision medicines, particularly when combined with nanotechnologies, holds potential as a way in which to overcome these obstacles and progress ferroptosis-inducing therapies into the clinic. Here, we review the current state-of-play of ferroptosis in haematological malignancies as well as encouraging discoveries in the field of ferroptosis nanotechnologies. While the research into ferroptosis nanotechnologies in haematological malignancies is limited, its pre-clinical success in solid tumours suggests this is a very feasible therapeutic approach to treat blood cancers such as multiple myeloma, lymphoma and leukaemia.
Collapse
Affiliation(s)
- Rachel L Mynott
- Flinders Health and Medical Research Institute, College of Medicine & Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Ali Habib
- Flinders Health and Medical Research Institute, College of Medicine & Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Oliver G Best
- Flinders Health and Medical Research Institute, College of Medicine & Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Craig T Wallington-Gates
- Flinders Health and Medical Research Institute, College of Medicine & Public Health, Flinders University, Adelaide, SA 5042, Australia
- Flinders Medical Centre, Bedford Park, SA 5042, Australia
| |
Collapse
|
135
|
Abstract
ABSTRACT Ferroptosis is an iron-dependent cell death pathway that is different from apoptosis, pyroptosis, and necrosis. The main characteristics of ferroptosis are the Fenton reaction mediated by intracellular free divalent iron ions, lipid peroxidation of cell membrane lipids, and inhibition of the anti-lipid peroxidation activity of intracellular glutathione peroxidase 4 (GPX4). Recent studies have shown that ferroptosis can be involved in the pathological processes of many disorders, such as ischemia-reperfusion injury, nervous system diseases, and blood diseases. However, the specific mechanisms by which ferroptosis participates in the occurrence and development of acute leukemia still need to be more fully and deeply studied. This article reviews the characteristics of ferroptosis and the regulatory mechanisms promoting or inhibiting ferroptosis. More importantly, it further discusses the role of ferroptosis in acute leukemia and predicts a change in treatment strategy brought about by increased knowledge of the role of ferroptosis in acute leukemia.
Collapse
Affiliation(s)
- Tianxin Lyu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Xudong Li
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
136
|
Yang J, Jin F, Li H, Shen Y, Shi W, Wang L, Zhong L, Wu G, Wu Q, Li Y. Identification of mitochondrial respiratory chain signature for predicting prognosis and immunotherapy response in stomach adenocarcinoma. Cancer Cell Int 2023; 23:69. [PMID: 37062830 PMCID: PMC10105960 DOI: 10.1186/s12935-023-02913-x] [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: 11/19/2022] [Accepted: 03/29/2023] [Indexed: 04/18/2023] Open
Abstract
Stomach adenocarcinoma (STAD) is the third leading cause of cancer-related deaths and the fifth most prevalent malignancy worldwide. Mitochondrial respiratory chain complexes play a crucial role in STAD pathogenesis. However, how mitochondrial respiratory chain complex genes (MRCCGs) affect the prognosis and tumor microenvironment in STAD remains unclear. In this study, we systematically analyzed genetic alterations and copy number variations of different expression densities of MRCCGs, based on 806 samples from two independent STAD cohorts. Then we employed the unsupervised clustering method to classify the samples into three expression patterns based on the prognostic MRCCG expressions, and found that they were involved in different biological pathways and correlated with the clinicopathological characteristics, immune cell infiltration, and prognosis of STAD. Subsequently, we conducted a univariate Cox regression analysis to identify the prognostic value of 1175 subtype-related differentially expressed genes (DEGs) and screened out 555 prognostic-related genes. Principal component analysis was performed and developed the MG score system to quantify MRCCG patterns of STAD. The prognostic significance of MG Score was validated in three cohorts. The low MG score group, characterized by increased microsatellite instability-high (MSI-H), tumor mutation burden (TMB), PD-L1 expression, had a better prognosis. Interestingly, we demonstrated MRCCG patterns score could predict the sensitivity to ferroptosis inducing therapy. Our comprehensive analysis of MRCCGs in STAD demonstrated their potential roles in the tumor-immune-stromal microenvironment, clinicopathological features, and prognosis. Our findings highlight that MRCCGs may provide a new understanding of immunotherapy strategies for gastric cancer and provide a new perspective on the development of personalized immune therapeutic strategies for patients with STAD.
Collapse
Affiliation(s)
- Jing Yang
- Laboratory Medicine Center, Department of Laboratory Medicine, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
- Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Feifan Jin
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huanjuan Li
- Laboratory Medicine Center, Department of Laboratory Medicine, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhuan Shen
- Laboratory Medicine Center, Department of Laboratory Medicine, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Weilin Shi
- Department of Medicine, Taizhou Luqiao District Second People's Hospital, Taizhou, Zhejiang, 318058, China
| | - Lina Wang
- Department of Medicine, Taizhou Luqiao District Second People's Hospital, Taizhou, Zhejiang, 318058, China
| | - Lei Zhong
- Department of Clinical Laboratory, Tongxiang Hospital of Traditional Chinese Medicine, Jiaxing, Zhejiang, 314599, China
| | - Gongqiang Wu
- Department of Hematology, Dongyang People's Hospital, Dongyang Hospital Affiliated to Wenzhou Medical University, Dongyang, Zhejiang, 322100, China.
| | - Qiaoliang Wu
- Department of Hematology, Jiashan first people's Hospital, Jiaxing, Zhejiang, 314199, China.
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
137
|
Liang J, Liao Y, Wang P, Yang K, Wang Y, Wang K, Zhong B, Zhou D, Cao Q, Li J, Zhao Y, Jiang N. Ferroptosis landscape in prostate cancer from molecular and metabolic perspective. Cell Death Discov 2023; 9:128. [PMID: 37061523 PMCID: PMC10105735 DOI: 10.1038/s41420-023-01430-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/17/2023] Open
Abstract
Prostate cancer is a major disease that threatens men's health. Its rapid progression, easy metastasis, and late castration resistance have brought obstacles to treatment. It is necessary to find new effective anticancer methods. Ferroptosis is a novel iron-dependent programmed cell death that plays a role in various cancers. Understanding how ferroptosis is regulated in prostate cancer will help us to use it as a new way to kill cancer cells. In this review, we summarize the regulation and role of ferroptosis in prostate cancer and the relationship with AR from the perspective of metabolism and molecular pathways. We also discuss the feasibility of ferroptosis in prostate cancer treatment and describe current limitations and prospects, providing a reference for future research and clinical application of ferroptosis.
Collapse
Affiliation(s)
- Jiaming Liang
- Tianjin institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Yihao Liao
- Tianjin institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Pu Wang
- Tianjin institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Kun Yang
- School of Future Technology, Xi'an Jiaotong University, 710049, Xi'an, Shaanxi, China
| | - Youzhi Wang
- Tianjin institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Keke Wang
- Department of Urology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Boqiang Zhong
- Tianjin institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Diansheng Zhou
- Tianjin institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Qian Cao
- Tianjin institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Junbo Li
- Tianjin institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Yang Zhao
- Department of Radiology, Tianjin Medical University Second Hospital, Tianjin, China
| | - Ning Jiang
- Tianjin institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China.
| |
Collapse
|
138
|
Akiyama H, Carter BZ, Andreeff M, Ishizawa J. Molecular Mechanisms of Ferroptosis and Updates of Ferroptosis Studies in Cancers and Leukemia. Cells 2023; 12:1128. [PMID: 37190037 PMCID: PMC10136912 DOI: 10.3390/cells12081128] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Ferroptosis is a mode of cell death regulated by iron-dependent lipid peroxidation. Growing evidence suggests ferroptosis induction as a novel anti-cancer modality that could potentially overcome therapy resistance in cancers. The molecular mechanisms involved in the regulation of ferroptosis are complex and highly dependent on context. Therefore, a comprehensive understanding of its execution and protection machinery in each tumor type is necessary for the implementation of this unique cell death mode to target individual cancers. Since most of the current evidence for ferroptosis regulation mechanisms is based on solid cancer studies, the knowledge of ferroptosis with regard to leukemia is largely lacking. In this review, we summarize the current understanding of ferroptosis-regulating mechanisms with respect to the metabolism of phospholipids and iron as well as major anti-oxidative pathways that protect cells from ferroptosis. We also highlight the diverse impact of p53, a master regulator of cell death and cellular metabolic processes, on the regulation of ferroptosis. Lastly, we discuss recent ferroptosis studies in leukemia and provide a future perspective for the development of promising anti-leukemia therapies implementing ferroptosis induction.
Collapse
Affiliation(s)
| | | | | | - Jo Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (H.A.); (B.Z.C.); (M.A.)
| |
Collapse
|
139
|
Shi H, Xiong L, Yan G, Du S, Liu J, Shi Y. Susceptibility of cervical cancer to dihydroartemisinin-induced ferritinophagy-dependent ferroptosis. Front Mol Biosci 2023; 10:1156062. [PMID: 37065442 PMCID: PMC10102504 DOI: 10.3389/fmolb.2023.1156062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
The clinical therapeutics of cervical cancer is limited due to the drug resistance and metastasis of tumor. As a novel target for antitumor therapy, ferroptosis is deemed to be more susceptible for those cancer cells with resistance to apoptosis and chemotherapy. Dihydroartemisinin (DHA), the primary active metabolites of artemisinin and its derivatives, has exhibited a variety of anticancer properties with low toxicity. However, the role of DHA and ferroptosis in cervical cancer remained unclear. Here, we showed that DHA could time-dependently and dose-dependently inhibit the proliferation of cervical cancer cells, which could be alleviated by the inhibitors of ferroptosis rather than apoptosis. Further investigation confirmed that DHA treatment initiated ferroptosis, as evidenced by the accumulation of reactive oxygen species (ROS), malondialdehyde (MDA) and liquid peroxidation (LPO) levels and simultaneously depletion of glutathione peroxidase 4 (GPX4) and glutathione (GSH). Moreover, nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy was also induced by DHA leading to subsequent increases of intracellular labile iron pool (LIP), exacerbated the Fenton reaction resulting in excessive ROS production, and enhanced cervical cancer ferroptosis. Among them, we unexpectedly found that heme oxygenase-1 (HO-1) played an antioxidant role in DHA-induced cell death. In addition, the results of synergy analysis showed that the combination of DHA and doxorubicin (DOX) emerged a highly synergistic lethal effect for cervical cancer cells, which was related also to ferroptosis. Overall, our data revealed the molecular mechanisms that DHA triggered ferritinophagy-dependent ferroptosis and sensitized to DOX in cervical cancer, which may provide novel avenues for future therapy development.
Collapse
Affiliation(s)
- Hanqiang Shi
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
| | - Lie Xiong
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
| | - Guang Yan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuqin Du
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Jiaxing, China
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Jie Liu
- Oncology Department, Jiaxing Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Jiaxing, China
| | - Yanbo Shi
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
- *Correspondence: Yanbo Shi,
| |
Collapse
|
140
|
Zhang R, Kang R, Tang D. Ferroptosis in gastrointestinal cancer: From mechanisms to implications. Cancer Lett 2023; 561:216147. [PMID: 36965540 DOI: 10.1016/j.canlet.2023.216147] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
Ferroptosis is a form of regulated cell death that is initiated by excessive lipid peroxidation that results in plasma membrane damage and the release of damage-associated molecular patterns. In recent years, ferroptosis has gained significant attention in cancer research due to its unique mechanism compared to other forms of regulated cell death, especially caspase-dependent apoptotic cell death. Gastrointestinal (GI) cancer encompasses malignancies that arise in the digestive tract, including the stomach, intestines, pancreas, colon, liver, rectum, anus, and biliary system. These cancers are a global health concern, with high incidence and mortality rates. Despite advances in medical treatments, drug resistance caused by defects in apoptotic pathways remains a persistent challenge in the management of GI cancer. Hence, exploring the role of ferroptosis in GI cancers may lead to more efficacious treatment strategies. In this review, we provide a comprehensive overview of the core mechanism of ferroptosis and discuss its function, regulation, and implications in the context of GI cancers.
Collapse
Affiliation(s)
- Ruoxi Zhang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
141
|
Zheng Y, Qin C, Li F, Qi J, Chu X, Li H, Shi T, Yan Z, Yang L, Xin X, Liu L, Han X, Yin L. Self-assembled thioether-bridged paclitaxel-dihydroartemisinin prodrug for amplified antitumor efficacy-based cancer ferroptotic-chemotherapy. Biomater Sci 2023; 11:3321-3334. [PMID: 36946490 DOI: 10.1039/d2bm02032g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Ferroptosis has been proposed as one form of iron-dependent cell death, overgeneration of high-toxicity hydroxyl radicals (˙OH) tumor sites via Fenton reactions induced cell membrane damage. However, the insufficient intracellular concentrations of both iron and H2O2 limited the anticancer performance of ferroptosis. In this study, ROS-sensitive prodrug nanoassemblies composed of a PEG2000-ferrous compound and a single thioether bond bridged dihydroartemisinin-paclitaxel prodrug were constructed, which fully tapped ex/endogenous iron, ferroptosis inducers, and chemotherapeutic agents. Following cellular uptake, the intracellular oxidizing environment accelerated the self-destruction of nanoassemblies and triggered drug release. In addition to the chemotherapeutic effect, the activated dihydroartemisinin was capable of acting as a toxic ˙OH amplifier via the reinforced Fenton reaction, simultaneously depleting intracellular GSH, as well as inducing glutathione peroxidase 4 inactivation, further enhancing ferroptosis-dependent cancer cell proliferation inhibition. Meanwhile, the ROS generation-inductive and cell cycle arrest effect from the paclitaxel augmented synergetic ferroptotic-chemotherapy of cancer. Thus, the prodrug integrating dihydroartemisinin with paclitaxel via a single thioether bond represents a potent nanoplatform to exert amplified ferroptotic-chemotherapy for improved anticancer efficacy.
Collapse
Affiliation(s)
- Yifei Zheng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Chao Qin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Fei Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Jingxin Qi
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Xinyu Chu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Ting Shi
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhen Yan
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Lei Yang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaofei Xin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Lisha Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaopeng Han
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Lifang Yin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, China; State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
142
|
Gong L, Huang D, Shi Y, Liang Z, Bu H. Regulated cell death in cancer: from pathogenesis to treatment. Chin Med J (Engl) 2023; 136:653-665. [PMID: 35950752 PMCID: PMC10129203 DOI: 10.1097/cm9.0000000000002239] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Regulated cell death (RCD), including apoptosis, pyroptosis, necroptosis, and ferroptosis, is regulated by a series of evolutionarily conserved pathways, and is required for development and tissue homeostasis. Based on previous genetic and biochemical explorations of cell death subroutines, the characteristics of each are generally considered distinctive. However, recent in-depth studies noted the presence of crosstalk between the different forms of RCD; hence, the concept of PANoptosis appeared. Cancer, a complex genetic disease, is characterized by stepwise deregulation of cell apoptosis and proliferation, with significant morbidity and mortality globally. At present, studies on the different RCD pathways, as well as the intricate relationships between different cell death subroutines, mainly focus on infectious diseases, and their roles in cancer remain unclear. As cancers are characterized by dysregulated cell death and inflammatory responses, most current treatment strategies aim to selectively induce cell death via different RCD pathways in cancer cells. In this review, we describe five types of RCD pathways in detail with respect to tumorigenesis and cancer progression. The potential value of some of these key effector molecules in tumor diagnosis and therapeutic response has also been raised. We then review and highlight recent progress in cancer treatment based on PANoptosis and ferroptosis induced by small-molecule compounds, immune checkpoint inhibitors, and nanoparticles. Together, these findings may provide meaningful evidence to fill in the gaps between cancer pathogenesis and RCD pathways to develop better cancer therapeutic strategies.
Collapse
Affiliation(s)
- Linjing Gong
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Dong Huang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yujun Shi
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zong’an Liang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hong Bu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
143
|
Grignano E, Cantero-Aguilar L, Tuerdi Z, Chabane T, Vazquez R, Johnson N, Zerbit J, Decroocq J, Birsen R, Fontenay M, Kosmider O, Chapuis N, Bouscary D. Dihydroartemisinin-induced ferroptosis in acute myeloid leukemia: links to iron metabolism and metallothionein. Cell Death Discov 2023; 9:97. [PMID: 36928207 PMCID: PMC10020442 DOI: 10.1038/s41420-023-01371-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 03/18/2023] Open
Abstract
Artemisinin is an anti-malarial drug that has shown anticancer properties. Recently, ferroptosis was reported to be induced by dihydroartemisinin (DHA) and linked to iron increase. In the current study, we determined the effect of DHA in leukemic cell lines on ferroptosis induction and iron metabolism and the cytoprotective effect triggered in leukemic cells. We found that treatment of DHA induces early ferroptosis by promoting ferritinophagy and subsequent iron increase. Furthermore, our study demonstrated that DHA activated zinc metabolism signaling, especially the upregulation of metallothionein (MT). Supportingly, we showed that inhibition MT2A and MT1M isoforms enhanced DHA-induced ferroptosis. Finally, we demonstrated that DHA-induced ferroptosis alters glutathione pool, which is highly dependent on MTs-driven antioxidant response. Taken together, our study indicated that DHA activates ferritinophagy and subsequent ferroptosis in AML and that MTs are involved in glutathione regenerating and antioxidant response.
Collapse
Affiliation(s)
- E Grignano
- INSERM U1016, Institut Cochin, Paris, France. .,CNRS UMR8104, Paris, France. .,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France. .,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France. .,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Clinique, Paris, France.
| | - L Cantero-Aguilar
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France
| | - Z Tuerdi
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France
| | - T Chabane
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France
| | - R Vazquez
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Biologique, Paris, France
| | - N Johnson
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Clinique, Paris, France
| | - J Zerbit
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Pharmacie, Paris, France
| | - J Decroocq
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Clinique, Paris, France
| | - R Birsen
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Clinique, Paris, France
| | - M Fontenay
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Biologique, Paris, France
| | - O Kosmider
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Biologique, Paris, France
| | - N Chapuis
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Biologique, Paris, France
| | - D Bouscary
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France.,Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Service d'Hématologie Clinique, Paris, France.,Member of OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France
| |
Collapse
|
144
|
Garciaz S, Miller T, Collette Y, Vey N. Targeting regulated cell death pathways in acute myeloid leukemia. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:151-168. [PMID: 37065864 PMCID: PMC10099605 DOI: 10.20517/cdr.2022.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 01/24/2023] [Accepted: 03/01/2023] [Indexed: 03/17/2023]
Abstract
The use of the BCL2 inhibitor venetoclax has transformed the management of patients with acute myeloid leukemia (AML) who are ineligible for intensive chemotherapy. By triggering intrinsic apoptosis, the drug is an excellent illustration of how our greater understanding of molecular cell death pathways can be translated into the clinic. Nevertheless, most venetoclax-treated patients will relapse, suggesting the need to target additional regulated cell death pathways. To highlight advances in this strategy, we review the recognized regulated cell death pathways, including apoptosis, necroptosis, ferroptosis and autophagy. Next, we detail the therapeutic opportunities to trigger regulated cell death in AML. Finally, we describe the main drug discovery challenges for regulated cell death inducers and their translation into clinical trials. A better knowledge of the molecular pathways regulating cell death represents a promising strategy to develop new drugs to cure resistant or refractory AML patients, particularly those resistant to intrinsic apoptosis.
Collapse
Affiliation(s)
- Sylvain Garciaz
- Hematology Department, Integrative Structural and Chemical Biology, Aix-Marseille Université, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Marseille 13009, France
| | - Thomas Miller
- Integrative Structural and Chemical Biology, Aix-Marseille Université, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Marseille 13009, France
| | - Yves Collette
- Integrative Structural and Chemical Biology, Aix-Marseille Université, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Marseille 13009, France
| | - Norbert Vey
- Hematology Department, Aix-Marseille Université, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), Marseille 13009, France
| |
Collapse
|
145
|
Abstract
Significance: Autophagy is a self-degrading process that determines cell fate in response to various environmental stresses. In contrast to autophagy-mediated cell survival, the signals, mechanisms, and effects of autophagy-dependent cell death remain obscure. The discovery of autophagy-dependent ferroptosis provides a paradigm for understanding the relationship between aberrant degradation pathways and excessive lipid peroxidation in driving regulated cell death. Recent Advances: Ferroptosis was originally described as an autophagy-independent and iron-mediated nonapoptotic cell death. Current studies reveal that the level of intracellular autophagy is positively correlated with ferroptosis sensitivity. Selective autophagic degradation of proteins (e.g., ferritin, SLC40A1, ARNTL, GPX4, and CDH2) or organelles (e.g., lipid droplets or mitochondria) promotes ferroptosis by inducing iron overload and/or lipid peroxidation. Several upstream autophagosome regulators (e.g., TMEM164), downstream autophagy receptors (e.g., HPCAL1), or danger signals (e.g., DCN) are selectively required for ferroptosis-related autophagy, but not for starvation-induced autophagy. The induction of autophagy-dependent ferroptosis is an effective approach to eliminate drug-resistant cancer cells. Critical Issues: How different organelles selectively activate autophagy to modulate ferroptosis sensitivity is not fully understood. Identifying direct protein effectors of ferroptotic cell death remains a challenge. Future Directions: Further understanding of the molecular mechanics and immune consequences of autophagy-dependent ferroptosis is critical for the development of precision antitumor therapies.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Xiutao Cai
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
146
|
Iron metabolism and ferroptosis in type 2 diabetes mellitus and complications: mechanisms and therapeutic opportunities. Cell Death Dis 2023; 14:186. [PMID: 36882414 PMCID: PMC9992652 DOI: 10.1038/s41419-023-05708-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 03/09/2023]
Abstract
The maintenance of iron homeostasis is essential for proper endocrine function. A growing body of evidence suggests that iron imbalance is a key factor in the development of several endocrine diseases. Nowadays, ferroptosis, an iron-dependent form of regulated cell death, has become increasingly recognized as an important process to mediate the pathogenesis and progression of type 2 diabetes mellitus (T2DM). It has been shown that ferroptosis in pancreas β cells leads to decreased insulin secretion; and ferroptosis in the liver, fat, and muscle induces insulin resistance. Understanding the mechanisms concerning the regulation of iron metabolism and ferroptosis in T2DM may lead to improved disease management. In this review, we summarized the connection between the metabolic pathways and molecular mechanisms of iron metabolism and ferroptosis in T2DM. Additionally, we discuss the potential targets and pathways concerning ferroptosis in treating T2DM and analysis the current limitations and future directions concerning these novel T2DM treatment targets.
Collapse
|
147
|
Sun SP, Lu YF, Li H, Weng CY, Chen JJ, Lou YJ, Lyu D, Lyu B. AMPK activation alleviated dextran sulfate sodium-induced colitis by inhibiting ferroptosis. J Dig Dis 2023; 24:213-223. [PMID: 37210607 DOI: 10.1111/1751-2980.13176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023]
Abstract
OBJECTIVES Ferroptosis is a newly discovered cell death mode that has been confirmed to occur in the intestinal epithelial cells in ulcerative colitis (UC). In this study we aimed to elucidate the mechanism of ferroptosis and its association with adenosine monophosphate-activated protein kinase (AMPK) in UC. METHODS Gene expression profiles of colonic mucosa (GSE87473) were downloaded. Both human colonic samples and dextran sodium sulfate (DSS)-induced colitis murine model were used. The molecular markers of ferroptosis were detected using western blot and immunohistochemistry. Symptoms, iron abundance, and lipid peroxidation level of the mouse model were measured to evaluate the role of AMPK activation in ferroptosis. RESULTS Both gene and protein expressions of GPX4 and FTH1 were decreased in UC patients compared with the healthy controls. An increased iron abundance and lipid peroxidation level in colon tissues and damaged mitochondria were found in DSS-induced colitis. AMPK expression was decreased in UC patients and correlated with FTH1 and GPX4. Activation of AMPK with metformin inhibited ferroptosis in the colon, improved symptoms, and prolonged the lifespan in DSS-induced colitis mice. CONCLUSIONS Ferroptosis can be observed in colonic tissues in UC. AMPK activation inhibits ferroptosis in murine colitis model, which may act as a potential target for the treatment of colitis.
Collapse
Affiliation(s)
- Shao Peng Sun
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi Fan Lu
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Heng Li
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Chun Yan Weng
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jia Jia Chen
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Yi Jie Lou
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Dong Lyu
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Bin Lyu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| |
Collapse
|
148
|
Wang Y, Ma Y, Jiang K. The role of ferroptosis in prostate cancer: a novel therapeutic strategy. Prostate Cancer Prostatic Dis 2023; 26:25-29. [PMID: 36056183 PMCID: PMC10023567 DOI: 10.1038/s41391-022-00583-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/26/2022] [Accepted: 08/15/2022] [Indexed: 11/09/2022]
Abstract
The incidence of prostate cancer is the second most among male cancers after lung cancer. Prostate cancer develops rapidly and is inclined to metastasize, and castration-resistant prostate cancer (CRPC) can be formed in the later stage, which brings great challenges to the prognosis and treatment. At present, the main treatment of prostate cancer is generally divided into four methods: surgery, chemotherapy, radiotherapy and endocrine therapy. However, the efficacy of these methods fails to satisfy the demands of patient prognosis. Ferroptosis is a newly discovered iron-dependent process, characterized by lipid peroxidation. Ferroptosis is associated with many diseases, especially tumor growth. In recent years, inhibiting tumor growth and overcoming tumor drug resistance by inducing ferroptosis has become a hot research topic. Previous studies have shown that induction of ferroptosis may be a new treatment for prostate cancer. We review the research progress of ferroptosis in prostate cancer in order to provide highly effective therapies for patients with prostate cancer.
Collapse
Affiliation(s)
- Yue Wang
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yifan Ma
- Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Kui Jiang
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
149
|
Abstract
Significance: The significance of ferroptosis in cancer therapeutics has now been unveiled. Specific ferroptosis inducers are expected as a promising strategy for cancer treatment, especially in cancers with epithelial mesenchymal transition and possibly in cancers with activated Hippo signaling pathways, both of which cause resistance to traditional chemotherapy but tend to show ferroptosis susceptibility. Recent Advances: Ferroptosis is a new form of regulated non-apoptotic cell death, which is characterized by iron-dependent lipid peroxidation, leading eventually to plasma membrane rupture. Its core mechanisms have been elucidated, consisting of a driving force as catalytic Fe(II)-dependent Fenton reaction and an incorporation of polyunsaturated fatty acids to membrane phospholipids via peroxisome-dependent and -independent pathways, and suppressing factors as prevention of lipid peroxidation with glutathione peroxidase 4 and direct membrane repair via coenzyme Q10 and ESCRT-III pathways. Critical Issues: Developments of ferroptosis inducers are in progress by nanotechnology-based drugs or by innovative engineering devices. Especially, low-temperature (non-thermal) plasma is a novel technology at the preclinical stage. The exposure can induce ferroptosis selectively in cancer cells rich in catalytic Fe(II). Future Directions: We also summarize and discuss the recently uncovered responsible molecular mechanisms in association with iron metabolism, ferroptosis and cancer therapeutics. Targeting ferroptosis in addition to the current therapeutic modalities would be important to cure advanced-stage cancer.
Collapse
Affiliation(s)
- Yashiro Motooka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Biosystem Science Division, Center for Low-Temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
150
|
Shi S, Luo H, Ji Y, Ouyang H, Wang Z, Wang X, Hu R, Wang L, Wang Y, Xia J, Cheng B, Bao B, Li X, Liao G, Xu B. Repurposing Dihydroartemisinin to Combat Oral Squamous Cell Carcinoma, Associated with Mitochondrial Dysfunction and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:9595201. [PMID: 37273554 PMCID: PMC10239307 DOI: 10.1155/2023/9595201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 08/15/2023]
Abstract
Oral squamous cell carcinoma (OSCC), with aggressive locoregional invasion, has a high rate of early recurrences and poor prognosis. Dihydroartemisinin (DHA), as a derivative of artemisinin, has been found to exert potent antitumor activity. Recent studies reported that DHA suppresses OSCC cell growth and viability through the regulation of reactive oxygen species (ROS) production and mitochondrial calcium uniporter. However, the mechanism underlying the action of DHA on OSCCs remains elusive. In the study, we observed that 159 genes were remarkably misregulated in primary OSCC tumors associated with DHA-inhibited pathways, supporting that OSCCs are susceptible to DHA treatment. Herein, our study showed that DHA exhibited promising effects to suppress OSCC cell growth and survival, and single-cell colony formation. Interestingly, the combination of DHA and cisplatin (CDDP) significantly reduced the toxicity of CDDP treatment alone on human normal oral cells (NOK). Moreover, DHA remarkably impaired mitochondrial structure and function, and triggered DNA damage and ROS generation, and activation of mitophagy. In addition, DHA induced leakage of cytochrome C and apoptosis-inducing factor (AIF) from mitochondria, elevated Bax/cleaved-caspase 3 expression levels and compromised Bcl2 protein expression. In the OSCC tumor-xenograft mice model, DHA remarkably suppressed tumor growth and induced apoptosis of OSCCs in vivo. Intriguingly, a selective mitophagy inhibitor Mdivi-1 could significantly reinforce the anticancer activity of DHA treatment. DHA and Mdivi-1 can synergistically suppress OSCC cell proliferation and survival. These data uncover a previously unappreciated contribution of the mitochondria-associated pathway to the antitumor activity of DHA on OSCCs. Our study shed light on a new aspect of a DHA-based therapeutic strategy to combat OSCC tumors.
Collapse
Affiliation(s)
- Shanwei Shi
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Hospital of Stomatology, Department of Oral and Maxillofacial Surgery, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Huigen Luo
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuna Ji
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Huiya Ouyang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zheng Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xinchen Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Renjie Hu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lihong Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yun Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Juan Xia
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Bin Cheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Baicheng Bao
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xin Li
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Guiqing Liao
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Hospital of Stomatology, Department of Oral and Maxillofacial Surgery, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Baoshan Xu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|