1
|
Chen S, Huang Y, Liu Y, Jiang L, Chen Y. Shikonin promotes ferroptosis though NSUN2-mediated m 5C methylation modification of TFRC in acute myelocytic leukemia. Hematology 2025; 30:2495221. [PMID: 40270446 DOI: 10.1080/16078454.2025.2495221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 04/12/2025] [Indexed: 04/25/2025] Open
Abstract
Shikonin (SHK), extracted from the traditional Chinese herb Lithospermum erythrorhizon, demonstrates a wide range of pharmacological activities. This study aimed to explore the role and underlying mechanisms of the 5-methylcytosine (m5C) RNA methyltransferase NOL1/NOP2/SUN domain (NSUN)2 in acute myelocytic leukemia (AML). To assess cell viability and death, we employed Cell Counting Kit-8 and propidium iodide staining. Ferroptosis-related markers were evaluated using commercial kits and Western blot analysis. The m5C levels of ferroptosis-associated mRNAs were quantified by methylated RNA immunoprecipitation (MeRIP)-qPCR. The specific m5C sites on the transferrin receptor (TFRC) mRNA were identified through a dual-luciferase reporter assay, while the interaction between NSUN2 and TFRC was investigated using RNA immunoprecipitation (RIP). The role of SHK in vivo was explored using a xenografted tumor model. Our findings revealed that SHK significantly reduced cell viability and induced cell death and ferroptosis in HL-60 and NB4 cells. Notably, SHK treatment led to an upregulation of NSUN2 expression. Inhibition of NSUN2 reversed the effects of SHK, restoring cell viability and reducing cell death and ferroptosis. Mechanistically, NSUN2 enhanced TFRC expression via m5C-dependent methylation. Overexpression of NSUN2 similarly decreased cell viability and increased cell death and ferroptosis, effects that were mitigated upon silencing of TFRC. In vivo, SHK treatment effectively suppressed tumor growth in xenografted mice. In summary, our study demonstrated that SHK promoted cell death and ferroptosis in AML by modulating NSUN2-mediated m5C methylation of TFRC. These findings provided novel insights into potential therapeutic strategies for AML.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Mice
- Animals
- Methyltransferases/metabolism
- Methyltransferases/genetics
- Receptors, Transferrin/metabolism
- Receptors, Transferrin/genetics
- Naphthoquinones/pharmacology
- Methylation
- Cell Line, Tumor
- HL-60 Cells
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Shuyu Chen
- School of Pharmacy, Zhangzhou Health Vocational College, Zhangzhou, People's Republic of China
| | - Yiqun Huang
- Department of Hematology, Zhangzhou Affiliated Hospital to Fujian Medical University, Zhangzhou, People's Republic of China
| | - Yinhao Liu
- Department of Diabetes Nephrology, Zhangzhou Hospital of Traditional Chinese Medicine, Zhangzhou, People's Republic of China
| | - Liuxuan Jiang
- The School of Clinical Medicine, Fujian Medical University, Zhangzhou, People's Republic of China
| | - Yuqing Chen
- School of Pharmacy, Zhangzhou Health Vocational College, Zhangzhou, People's Republic of China
| |
Collapse
|
2
|
Liu S, Yang X, Zheng S, Chen C, Qi L, Xu X, Zhang D. Research progress on the use of traditional Chinese medicine to treat diseases by regulating ferroptosis. Genes Dis 2025; 12:101451. [PMID: 40070365 PMCID: PMC11894312 DOI: 10.1016/j.gendis.2024.101451] [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: 01/28/2024] [Revised: 06/28/2024] [Accepted: 08/25/2024] [Indexed: 03/14/2025] Open
Abstract
Ferroptosis is an emerging form of programmed cell death triggered by iron-dependent lipid peroxidation. It is distinguished from other forms of cell death by its unique morphological changes and characteristic fine-tuned regulatory gene network. Since its pivotal involvement in the pathogenesis and therapeutic interventions of various diseases, such as malignant tumors, cardiovascular and cerebrovascular diseases, and traumatic disorders, has been well-established, ferroptosis has garnered significant attention in contemporary physiological and pathological research. For the advantage of alleviating the clinical symptoms and improving life quality, traditional Chinese medicine (TCM) holds a significant position in the treatment of these ailments. Moreover, increasing studies revealed that TCM compounds and monomers showed evident therapeutic efficacy by regulating ferroptosis via signaling pathways that tightly regulate redox reactions, iron ion homeostasis, lipid peroxidation, and glutathione metabolism. In this paper, we summarized the current knowledge of TCM compounds and monomers in regulating ferroptosis, aiming to provide a comprehensive review of disease management by TCM decoction, Chinese patent medicine, and natural products deriving from TCM through ferroptosis modulation. The formulation composition, chemical structure, and possible targets or mechanisms presented here offer valuable insights into the advancement of TCM exploration.
Collapse
Affiliation(s)
- Shuai Liu
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
- Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Xianzhen Yang
- Urinary Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Sanxia Zheng
- Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Changjing Chen
- Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Lei Qi
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
- Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Xiangdong Xu
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
- Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Denglu Zhang
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
- Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| |
Collapse
|
3
|
Xu J, Ren F, Wang J, Liu J, Cui X, Hao J, Yang W, Zhang Y, Cao D, Li L, Wang H. Tubeimoside I induces mitophagy by activating the PINK1/Parkin/Mfn2 signaling pathway in acute myeloid leukemia cells. Transl Oncol 2025; 55:102355. [PMID: 40112502 PMCID: PMC11979407 DOI: 10.1016/j.tranon.2025.102355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Acute myeloid leukemia (AML) is the most prevalent kind of acute leukemia in adults. Despite the availability of new targeted therapies, AML remains connected with a poor prognosis and decreased rate of survival. Tubeimoside I (TBMS1), a critical compound extracted from Bolbostemma paniculatum, has demonstrated potential anticancer effects in lung and colorectal cancers. Nevertheless, the TBMS1 anticancer pathway against AML is still elusive. This study aimed to explore the potential role of TBMS1 in anti-AML and its molecular mechanism. In vitro, TBMS1 treatment suppressed AML cells proliferation, induced apoptosis, and mitochondrial damage, and elevated ROS levels. Network pharmacological analysis suggested, and subsequent studies confirmed, that TBMS1 induced mitophagy in AML cells by modulating the PINK1/Parkin/Mfnh2 signaling pathway, an effect that was effectively reversed following PINK1 knockdown. In vivo, TBMS1 treatment suppressed the proliferation of AML cells after 21 days, improved the survival rates of nude mice, and showed no evident organ toxicity. These evidences suggest that TBMS1 may have significant therapeutic potential in treating AML.
Collapse
Affiliation(s)
- Jing Xu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan 030001, China
| | - Fanggang Ren
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Jinjuan Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Jianbing Liu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaohua Cui
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Jianqing Hao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Wanfang Yang
- School of Basic Medicine, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Yaofang Zhang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Dongmin Cao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Zhongshan 528437, China.
| | - Li Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan 030001, China.
| | - Hongwei Wang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China; School of Basic Medicine, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| |
Collapse
|
4
|
Zhao Y, Lu L, Chen X, Yin Q. Natural compounds targeting ferroptosis in ovarian cancer: Research progress and application potential. Pharmacol Res 2025; 215:107729. [PMID: 40194611 DOI: 10.1016/j.phrs.2025.107729] [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: 11/14/2024] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
Ovarian cancer (OC) is among the most common malignancies in the female reproductive system, marked by high rates of recurrence and mortality. Conventional chemotherapy, however, faces limitations due to the development of drug resistance, which hinders its effectiveness. Ferroptosis, an atypical form of programmed cell death distinct from autophagy, apoptosis, and necrosis, the relationship with tumors has become a hot research area in cancer studies in recent years. Anticancer therapies that target ferroptosis show strong potential in improving prognosis and counteracting chemotherapy resistance. Natural compounds, as a valuable source of novel targeted anticancer agents, its significant role in inhibiting tumor cell proliferation and metastasis and improving therapeutic sensitivity has been demonstrated in numerous existing studies. This review summarizes a range of natural compounds that target ferroptosis in OC cells, discussing their active components, mechanisms of action, and therapeutic potential, thereby providing useful insights for future targeted therapy and research in OC.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.
| | - Lichao Lu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China.
| | - Xingying Chen
- Yuebei People's Hospital, Shaoguan, Guangdong 512000, China.
| | - Qiaozhi Yin
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.
| |
Collapse
|
5
|
Zheng H, Lin Z, Wang D, Zhang J, Zeng T, Shen J, Li JD, Yang M. BMAL1-depletion remodels ceramide metabolism to regulate ferroptosis and sorafenib chemosensitivity in acute myeloid leukemia. iScience 2025; 28:112054. [PMID: 40241743 PMCID: PMC12002609 DOI: 10.1016/j.isci.2025.112054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/17/2024] [Accepted: 02/13/2025] [Indexed: 04/18/2025] Open
Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy with a poor prognosis. We discovered that BMAL1 is a ferroptosis suppressor. Furthermore, it was also found to be overexpressed in AML patients, affecting the cell cycle and promoting tumor cell growth and progression. In this study, we further validated the association of BMAL1 with the progression and survival outcomes of AML. Lipidomic revealed that the levels of ceramide increased in AML cells following the depletion of BMAL1. Ceramide facilitated ferroptosis in AML cells. ASAH2 played a key role in this process. BMAL1 could not directly regulate ASAH2 but instead through IKZF2. Elevated levels of ceramide promoted the degradation of the ferroptosis protection molecule GPX4, ultimately promoting ferroptosis. Furthermore, ceramide treatment has been demonstrated to enhance the responsiveness of AML cells to sorafenib. In summary, this study elucidates that BMAL1 depletion remodels ceramide metabolism to regulate the sensitivity of AML cells to ferroptosis and targeted drug sorafenib.
Collapse
Affiliation(s)
- Hong Zheng
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Department of Pediatrics, The Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Clinical Research Center of Pediatric Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- MOE Key Lab of Rare Pediatric Diseases, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Zhi Lin
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Hunan Clinical Research Center of Pediatric Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- MOE Key Lab of Rare Pediatric Diseases, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Dan Wang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Hunan Clinical Research Center of Pediatric Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- MOE Key Lab of Rare Pediatric Diseases, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Jing Zhang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
| | - Ting Zeng
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Department of Pediatrics, The Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Clinical Research Center of Pediatric Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- MOE Key Lab of Rare Pediatric Diseases, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Jie Shen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Hunan Clinical Research Center of Pediatric Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- MOE Key Lab of Rare Pediatric Diseases, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Jia-Da Li
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
| | - Minghua Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Hunan Clinical Research Center of Pediatric Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- MOE Key Lab of Rare Pediatric Diseases, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
6
|
Li F, Chen Y, Zhuang H, Pei R, Lu Y, Chen D, Li S, Ye P, Lian J, Lu Y. A combination of Dihydroartemisinin and Venetoclax enhances antitumor effect in AML via C-MYC/BCL-XL/MCL-1 triple targeting. Discov Oncol 2025; 16:496. [PMID: 40202582 PMCID: PMC11982003 DOI: 10.1007/s12672-025-02242-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is associated with high rates of resistance to standard therapies, necessitating the exploration of novel treatment strategies. Venetoclax (VEN) has shown efficacy in AML, yet drug resistance remains a significant challenge. This study aims to explore the synergistic effects of combining dihydroartemisinin (DHA) with VEN to improve therapeutic outcomes in AML. METHODS AML cell lines and primary cells from AML patients were treated with various concentrations of DHA, VEN and their combined regimen. The cytotoxic effects were evaluated using MTS assays, flow cytometry for apoptosis analysis, and cell cycle assessments. Protein levels of caspase-3, PARP, MCL-1, BCL-XL and C-MYC were analyzed to elucidate the underlying mechanisms of the observed synergy. RESULTS The combination of VEN and DHA demonstrated a significant synergistic cytotoxic effect on AML cells, characterized by reduced cell proliferation, induced apoptosis, and cell cycle arrest in the G0/G1 phase. Mechanistically, the synergy was associated with increased levels of cleaved caspase-3 and PARP, along with the downregulation of anti-apoptotic proteins MCL-1 and BCL-XL. Additionally, the combined treatment led to a significant decrease in C-MYC expression. This synergistic effect was consistently observed across all primary AML patient samples analyzed. CONCLUSION The findings suggest that the combination of VEN and DHA exerts synergistic anti-leukemic effects by targeting BCL-XL, MCL-1 and C-MYC, offering a promising therapeutic strategy for AML.
Collapse
Affiliation(s)
- Fenglin Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Yao Chen
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Shaoxing Central Hospital, Shaoxing, China
| | - Haihui Zhuang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Renzhi Pei
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Yuyu Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Dong Chen
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Shuangyue Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Peipei Ye
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Jiaying Lian
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Ying Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Baizhang Road 251#, Ningbo, China.
- Institute of Hematology, Ningbo University, Ningbo, China.
| |
Collapse
|
7
|
Mo H, Fang H, Jia L, Zhou S, Feng M, Wu X, Yuan W. Enhancing sensitivity to oxaliplatin in tongue squamous cell carcinoma: mechanistic insights and therapeutic potential of DHA combination therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4393-4407. [PMID: 39476243 DOI: 10.1007/s00210-024-03548-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/17/2024] [Indexed: 04/10/2025]
Abstract
Squamous cell carcinoma of the tongue, a common and aggressive malignancy, poses a substantial threat to health and well-being. Despite the promising results of combination therapy with dihydroartemisinin (DHA) and oxaliplatin (Oxa) in various cancers, its effectiveness in treating tongue squamous cell carcinoma had not been explored prior to this study. Our research found that DHA significantly enhances the sensitivity of tongue squamous cell carcinoma cells to Oxa, even at very low concentrations. The combination treatment was observed to modulate the activity of CDK1 and Cyclin B1, arresting the cell cycle in the G2 phase. Additionally, it reduces mitochondrial membrane potential, prompting the release of cytochrome c and activating cleaved caspase-3, which promotes apoptosis. Notably, surface plasmon resonance and immunoprecipitation experiments revealed that DHA targets and attenuates CDK1 modification, weakening its interaction with STAT3 protein. This leads to reduced expression of anti-apoptotic genes and facilitates programmed cell death in CAL-27 cells. The findings underscore the potential of DHA and Oxa as a potent therapeutic strategy for tongue squamous cell carcinoma, opening avenues for clinical application and further exploration into its mechanistic pathways.
Collapse
Affiliation(s)
- Hailan Mo
- Chongqing Medical University, Chongqing, 400016, China
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing University, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, 401147, China
| | - Hongyan Fang
- Chongqing Medical University, Chongqing, 400016, China
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing University, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, 401147, China
| | - Lifeng Jia
- Chongqing Medical University, Chongqing, 400016, China
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing University, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, 401147, China
| | - Shitong Zhou
- Chongqing Medical University, Chongqing, 400016, China
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing University, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, 401147, China
| | - Menglong Feng
- Chongqing Medical University, Chongqing, 400016, China
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing University, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, 401147, China
| | - Xiaolu Wu
- Chongqing Medical University, Chongqing, 400016, China
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing University, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, 401147, China
| | - Wei Yuan
- Chongqing Medical University, Chongqing, 400016, China.
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing University, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, 401147, China.
| |
Collapse
|
8
|
Czechowicz P, Więch-Walów A, Sławski J, Collawn JF, Bartoszewski R. Old drugs, new challenges: reassigning drugs for cancer therapies. Cell Mol Biol Lett 2025; 30:27. [PMID: 40038587 DOI: 10.1186/s11658-025-00710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
The "War on Cancer" began with the National Cancer Act of 1971 and despite more than 50 years of effort and numerous successes, there still remains much more work to be done. The major challenge remains the complexity and intrinsic polygenicity of neoplastic diseases. Furthermore, the safety of the antitumor therapies still remains a concern given their often off-target effects. Although the amount of money invested in research and development required to introduce a novel FDA-approved drug has continuously increased, the likelihood for a new cancer drug's approval remains limited. One interesting alternative approach, however, is the idea of repurposing of old drugs, which is both faster and less costly than developing new drugs. Repurposed drugs have the potential to address the shortage of new drugs with the added benefit that the safety concerns are already established. That being said, their interactions with other new drugs in combination therapies, however, should be tested. In this review, we discuss the history of repurposed drugs, some successes and failures, as well as the multiple challenges and obstacles that need to be addressed in order to enhance repurposed drugs' potential for new cancer therapies.
Collapse
Affiliation(s)
- Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Anna Więch-Walów
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland.
| |
Collapse
|
9
|
Safari MH, Rahimzadeh P, Alaei E, Alimohammadi M, Esfandiari N, Daneshi S, Malgard N, Farahani N, Taheriazam A, Hashemi M. Targeting ferroptosis in gastrointestinal tumors: Interplay of iron-dependent cell death and autophagy. Mol Cell Probes 2025; 79:102013. [PMID: 39837469 DOI: 10.1016/j.mcp.2025.102013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
Ferroptosis is a regulated cell death mechanism distinct from apoptosis, autophagy, and necroptosis, marked by iron accumulation and lipid peroxidation. Since its identification in 2012, it has developed into a potential therapeutic target, especially concerning GI disorders like PC, HCC, GC, and CRC. This interest arises from the distinctive role of ferroptosis in the progression of diseases, presenting a new avenue for treatment where existing therapies fall short. Recent studies emphasize the promise of focusing on ferroptosis to fight GI cancers, showcasing its unique pathophysiological mechanisms compared to other types of cell death. By comprehending how ferroptosis aids in the onset and advancement of GI diseases, scientists aim to discover novel drug targets and treatment approaches. Investigating ferroptosis in gastrointestinal disorders reveals exciting possibilities for novel therapies, potentially revolutionizing cancer treatment and providing renewed hope for individuals affected by these tumors.
Collapse
Affiliation(s)
- Mohamad Hosein Safari
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Negin Esfandiari
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Neda Malgard
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
10
|
Sun W, Xia Y, Jin F, Cao J, Wu G, Li K, Yu Y, Wu Y, Ye G, Xu K, Liu D, Jin W. Identification of fatty acid anabolism patterns to predict prognosis and immunotherapy response in gastric cancer. Discov Oncol 2025; 16:6. [PMID: 39755916 DOI: 10.1007/s12672-025-01745-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/02/2025] [Indexed: 01/06/2025] Open
Abstract
Gastric cancer (GC), one of the most common and heterogeneous malignancies, is the second leading cause of cancer death worldwide and is closely related to dietary habits. Fatty acid is one of the main nutrients of human beings, which is closely related to diabetes, hypertension and other diseases. However, the correlation between fatty acid metabolism and the development and progression of GC remains largely unknown. Here, we profiled the genetic alterations of fatty acid anabolism-related genes (FARGs) in gastric cancer samples from the TCGA cohort and GEO database to evaluate the possible relationships and their internal regulatory mechanism. Through consistent clustering and functional enrichment analysis, three distinct fatty acid anabolism clusters and three gene subtypes were identified to participate in different biological pathways, and correlated with the characteristics of immune cell infiltration and clinical prognosis. Importantly, a distinctive FA-score was constructed through the principal component analysis to quantify the characteristics of fatty acid anabolism in each GC patient. Further analysis showed patients grouped in the high FA-score group were characterized with greater tumor mutational burden (TMB) and higher microsatellite stability (MSI-H), which may be more aeschynomenous to immunotherapy and had a favorable prognosis. Altogether, our bioinformatics analysis based on FARGs uncovered the potential roles of fatty acid metabolism in GC, and may provide newly prognostic information and novel approaches for promoting individualized immunotherapy in patients with GC.
Collapse
Affiliation(s)
- Weijie Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yanhong Xia
- Procurement Center, Hangzhou Children's Hospital, Hangzhou, Zhejiang, China
| | - Feifan Jin
- Department of Stomatology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Jinghao Cao
- Department of Clinical Laboratory, Laboratory Medicine Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Gaoping Wu
- Department of Clinical Laboratory, Adicon Clinical Laboratories, Inc., Hangzhou, Zhejiang, China
| | - Keyi Li
- Department of Clinical Laboratory, Laboratory Medicine Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanhua Yu
- Department of Clinical Laboratory, Laboratory Medicine Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yunyi Wu
- Department of Clinical Laboratory, Laboratory Medicine Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Gaoqi Ye
- Department of Clinical Laboratory, Laboratory Medicine Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ke Xu
- Department of Clinical Laboratory, Laboratory Medicine Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Dengpan Liu
- Department of Clinical Laboratory, Zhejiang University Sir Run Run Shaw Alaer Hospital, Alaer, Xinjiang, China.
| | - Weidong Jin
- Department of Clinical Laboratory, Laboratory Medicine Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
11
|
Xie Z, Hou Q, He Y, Xie Y, Mo Q, Wang Z, Zhao Z, Chen X, Peng T, Li L, Xie W. Ferritin Hinders Ferroptosis in Non-Tumorous Diseases: Regulatory Mechanisms and Potential Consequences. Curr Protein Pept Sci 2025; 26:89-104. [PMID: 39225224 DOI: 10.2174/0113892037315874240826112422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Ferritin, as an iron storage protein, has the potential to inhibit ferroptosis by reducing excess intracellular free iron concentrations and lipid reactive oxygen species (ROS). An insufficient amount of ferritin is one of the conditions that can lead to ferroptosis through the Fenton reaction mediated by ferrous iron. Consequently, upregulation of ferritin at the transcriptional or posttranscriptional level may inhibit ferroptosis. In this review, we have discussed the essential role of ferritin in ferroptosis and the regulatory mechanism of ferroptosis in ferritin-deficient individuals. The description of the regulatory factors governing ferritin and its properties in regulating ferroptosis as underlying mechanisms for the pathologies of diseases will allow potential therapeutic approaches to be developed.
Collapse
Affiliation(s)
- Zhongcheng Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Qin Hou
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yinling He
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yushu Xie
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Qinger Mo
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Ziyi Wang
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Ziye Zhao
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Liang Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
12
|
Niu X, You Q, Hou K, Tian Y, Wei P, Zhu Y, Gao B, Ashrafizadeh M, Aref AR, Kalbasi A, Cañadas I, Sethi G, Tergaonkar V, Wang L, Lin Y, Kang D, Klionsky DJ. Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist Updat 2025; 78:101170. [PMID: 39603146 DOI: 10.1016/j.drup.2024.101170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Macroautophagy/autophagy is a highly conserved evolutionary mechanism involving lysosomes for the degradation of cytoplasmic components including organelles. The constitutive, basal level of autophagy is fundamental for preserving cellular homeostasis; however, alterations in autophagy can cause disease pathogenesis, including cancer. The role of autophagy in cancer is particularly complicated, since this process acts both as a tumor suppressor in precancerous stages but facilitates tumor progression during carcinogenesis and later stages of cancer progression. This shift between anti-tumor and pro-tumor roles may be influenced by genetic and environmental factors modulating key pathways such as those involving autophagy-related proteins, the PI3K-AKT-MTOR axis, and AMPK, which often show dysregulation in tumors. Autophagy regulates various cellular functions, including metabolism of glucose, glutamine, and lipids, cell proliferation, metastasis, and several types of cell death (apoptosis, ferroptosis, necroptosis and immunogenic cell death). These multifaceted roles demonstrate the potential of autophagy to affect DNA damage repair, cell death pathways, proliferation and survival, which are critical in determining cancer cells' response to chemotherapy. Therefore, targeting autophagy pathways presents a promising strategy to combat chemoresistance, as one of the major reasons for the failure in cancer patient treatment. Furthermore, autophagy modulates immune evasion and the function of immune cells such as T cells and dendritic cells, influencing the tumor microenvironment and cancer's biological behavior. However, the therapeutic targeting of autophagy is complex due to its dual role in promoting survival and inducing cell death in cancer cells, highlighting the need for strategies that consider both the beneficial and detrimental effects of autophagy modulation in cancer therapy. Hence, both inducers and inhibitors of autophagy have been introduced for the treatment of cancer. This review emphasizes the intricate interplay between autophagy, tumor biology, and immune responses, offering insights into potential therapeutic approaches that deploy autophagy in the cancer suppression.
Collapse
Affiliation(s)
- Xuegang Niu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Qi You
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Kaijian Hou
- School of Public Health(Long Hu people hospital), Shantou University, Shantou, 515000, Guangdong, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, IL 60532, USA
| | - Penghui Wei
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Bin Gao
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Alireza Kalbasi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Israel Cañadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A⁎STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Lingzhi Wang
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Yuanxiang Lin
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Dezhi Kang
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Chen H, Lyu F, Gao X. Advances in ferroptosis for castration-resistant prostate cancer treatment: novel drug targets and combination therapy strategies. Prostate Cancer Prostatic Dis 2024:10.1038/s41391-024-00933-w. [PMID: 39733054 DOI: 10.1038/s41391-024-00933-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Metastatic prostate cancer (PCa) has much lower survival and ultimately develops castration resistance, which expects novel targets and therapeutic approaches. As a result of iron-dependent lipid peroxidation, ferroptosis triggers programmed cell death and has been associated with castration-resistant prostate cancer (CRPC). SUBJECTS To better understand how ferroptosis can be used to treat CRPC, we reviewed the following: First, ferroptosis mechanisms and characteristics. We then pay attention to ferroptosis effects on CRPC, and the relationship between ferroptosis and CRPC treatment. Finally, we'd like to figure out if ferroptosis could predict the prognosis of CRPC thus screening early for populations that may benefit from appropriate therapies. RESULTS The review demonstrated that ferroptosis regulators like PI3K/AKT/mTOR, DECR1 et al., have a significant role in the development of CRPC and that several inducers of ferroptosis, such as erastin, BSO, RSL3, and FIN56, have already demonstrated their effects in that area. What's more, ferroptosis is crucial for radiation-induced anticancer effects by inducing lipid peroxidation and regulating p53, AMPK, and others. Additionally, it has been discovered that certain GPX4 and SLC7A11 inhibitors can increase radiosensitivity, which brings new combination strategies. Finally, among the genes associated with ferroptosis, which may be excellent predictors of prostate cancer prognosis, several risk models have been developed and shown promising predictive capabilities. CONCLUSIONS Ferroptosis can serve as a potential therapeutic target for CRPC, and could be a new strategy for combination therapy. Moreover, ferroptosis-related genes may be great indicators of PCa prognosis. Further research on ferroptosis in CRPC therapy can benefit from the frameworks provided by this review.
Collapse
Affiliation(s)
- Huizhu Chen
- Department of Radiation Oncology, Peking University First Hospital, 100034, Beijing, China
| | - Feng Lyu
- Department of Radiation Oncology, Peking University First Hospital, 100034, Beijing, China
| | - Xianshu Gao
- Department of Radiation Oncology, Peking University First Hospital, 100034, Beijing, China.
| |
Collapse
|
14
|
Wang Y, Feng W, Wang F, Min J. [Research progress of iron metabolism and ferroptosis in myeloid neoplasms]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:735-746. [PMID: 39608794 PMCID: PMC11736352 DOI: 10.3724/zdxbyxb-2024-0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/05/2024] [Indexed: 11/30/2024]
Abstract
It is reported that iron metabolism and ferroptosis can influence the occurrence and development of myeloid tumors, which can serve as therapeutic targets. Dysregulation of iron metabolism is present in a variety of myeloid neoplasms. The prognosis of acute myeloid leukemia is related to differential expression of molecules related to iron metabolism. The prognosis of myelodysplastic syndrome patients with iron overload is poor. Myeloproliferative neoplasms are often characterized by the coexistence of iron deficiency and erythrocytosis, which can be treated by targeting hepcidin. Myeloid tumor cells are susceptible to oxidative damage caused by the accumulation of reactive oxygen species and are sensitive to ferroptosis. Ferroptosis has anti-tumor effect in acute myeloid leukemia and myelodysplastic syndrome. Targeting ferroptosis can reverse imatinib resistance in chronic myeloid leukemia. This article reviews the characteristics of iron metabolism in the development and progression of myeloid neoplasms, as well as the mechanism of ferroptosis, to provide a basis for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Yudi Wang
- Department of Hematology, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang Province, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Weiying Feng
- Department of Hematology, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Fudi Wang
- School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Junxia Min
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
15
|
Yang G, Yang S, Li J, Jiang P, Tian X, Wang X, Wei J, Zhang X, Liu J. Low-dose treatment with Epirubicin, a novel histone deacetylase 1 inhibitor, exerts anti-leukemic effects by inducing ferroptosis. Eur J Pharmacol 2024; 985:177058. [PMID: 39413949 DOI: 10.1016/j.ejphar.2024.177058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
AIMS Leukemia is hematopoietic stem cell malignant tumor with poor outcomes. Histone deacetylase 1 (HDAC1) is highly expressed in leukemia and current HDAC1 inhibitors have clinical limitations in leukemia therapy. Therefore, novel HDAC1 inhibitor is imperative to being found and its mechanism needs to be further explored. MATERIALS AND METHODS Novel HDAC1 inhibitors were discovered through drug virtual screening. CCK-8, EdU and soft agar assay were used to assess the anti-leukemic effect of the candidate HDAC1 inhibitor. ROS, lipid peroxidation, intracellular Fe2+ and LIP assay were employed to verify cell ferroptosis. Additionally, a xenograft model was performed to explore the efficacy and safety of the candidate HDAC1 inhibitor in vivo. RESULTS HDAC1 might be a promising therapeutic target for leukemia and Epirubicin (Epi) could be used as a potential HDAC1 inhibitor. Low-dose Epi exhibited good anti-leukemic effects by inhibiting cell proliferation, DNA synthesis and colony formation. Low-dose Epi could induce ferroptosis by triggering lipid peroxidation, which was better than that treated with current HDAC1 inhibitors Chidamide or Vorinostat, ROS generation and Fe2+ overload in leukemia cells. Mechanistically, low-dose Epi induced ferroptosis by targeting amino acid metabolism and iron metabolism. Similar results were found in a xenograft model in NOG mice with a good safety profile. CONCLUSION Our study demonstrated that Epi might be used as a HDAC1 inhibitor. Low-dose Epi could inhibit tumor progression by inducing cell ferroptosis in vitro and in vivo. Thus, Epi administration with lower concentration may be much more favorable and safer in the treatment with leukemia.
Collapse
Affiliation(s)
- Guancui Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China; Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Shijie Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jiarun Li
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Peijie Jiang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China; Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Xiaolong Tian
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xiaoqi Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jin Wei
- Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637002, China
| | - Xi Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 401329, China.
| | - Jinyi Liu
- State Key Laboratory of Trauma and Chemical Poisoning, Medical Center of Hematology, Military Key Clinical Specialty, Chongqing Key Clinical Specialty, Chongqing Key Laboratory of Hematology and Microenvironment, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| |
Collapse
|
16
|
Zhu K, Cai Y, Lan L, Luo N. Tumor Metabolic Reprogramming and Ferroptosis: The Impact of Glucose, Protein, and Lipid Metabolism. Int J Mol Sci 2024; 25:13413. [PMID: 39769177 PMCID: PMC11676715 DOI: 10.3390/ijms252413413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 01/03/2025] Open
Abstract
Ferroptosis, a novel form of cell death discovered in recent years, is typically accompanied by significant iron accumulation and lipid peroxidation during the process. This article systematically elucidates how tumor metabolic reprogramming affects the ferroptosis process in tumor cells. The paper outlines the basic concepts and physiological significance of tumor metabolic reprogramming and ferroptosis, and delves into the specific regulatory mechanisms of glucose metabolism, protein metabolism, and lipid metabolism on ferroptosis. We also explore how complex metabolic changes in the tumor microenvironment further influence the response of tumor cells to ferroptosis. Glucose metabolism modulates ferroptosis sensitivity by influencing intracellular energetic status and redox balance; protein metabolism, involving amino acid metabolism and protein synthesis, plays a crucial role in the initiation and progression of ferroptosis; and the relationship between lipid metabolism and ferroptosis primarily manifests in the generation and elimination of lipid peroxides. This review aims to provide a new perspective on how tumor cells regulate ferroptosis through metabolic reprogramming, with the ultimate goal of offering a theoretical basis for developing novel therapeutic strategies targeting tumor metabolism and ferroptosis.
Collapse
Affiliation(s)
- Keyu Zhu
- School of Medicine, Nankai University, Tianjin 300071, China; (K.Z.); (Y.C.)
| | - Yuang Cai
- School of Medicine, Nankai University, Tianjin 300071, China; (K.Z.); (Y.C.)
| | - Lan Lan
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China;
| | - Na Luo
- School of Medicine, Nankai University, Tianjin 300071, China; (K.Z.); (Y.C.)
| |
Collapse
|
17
|
Lyu H, Kong J, Chen J, Zhang R, Xiao S, Guo D, Zhang Q, Chen XZ, Tang J, Zhou C. The Emerging Scenario of Ferroptosis in Pancreatic Cancer Tumorigenesis and Treatment. Int J Mol Sci 2024; 25:13334. [PMID: 39769097 PMCID: PMC11727763 DOI: 10.3390/ijms252413334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 01/05/2025] Open
Abstract
Pancreatic cancer remains one of the most lethal forms of cancer. Currently, there is a lack of effective drug treatments for pancreatic cancer. However, as a newly discovered form of non-apoptotic cell death, ferroptosis has garnered increasing attention in relation to pancreatic cancer. Understanding the role of ferroptosis in the tumorigenesis and treatment of pancreatic cancer may enable more effective clinical trials and treatments for pancreatic cancer and may minimize side effects or restrict the emergence of drug resistance. In this review, we summarize the current knowledge regarding the process and underlying mechanisms of ferroptosis, as well as its dual role in both promoting tumorigenesis and facilitating treatment strategies for pancreatic cancer. Additionally, how ferroptosis is implicated in the development of pancreatitis and insulin resistance, indicating that ferroptosis may play an important role in the risk of pancreatitis- and insulin-resistance-related pancreatic cancers, is also addressed.
Collapse
Affiliation(s)
- Hao Lyu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Jinghua Kong
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Jiasi Chen
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Rui Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Shuai Xiao
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Dong Guo
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Qi Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Cefan Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
18
|
Zhang W, Wen W, Tan R, Zhang M, Zhong T, Wang J, Chen H, Fang X. Ferroptosis: Potential therapeutic targets and prognostic predictions for acute myeloid leukemia (Review). Oncol Lett 2024; 28:574. [PMID: 39397802 PMCID: PMC11467844 DOI: 10.3892/ol.2024.14707] [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: 06/05/2024] [Accepted: 08/15/2024] [Indexed: 10/15/2024] Open
Abstract
Ferroptosis is a relatively recently discovered type of regulated cell death that is induced by iron-dependent lipid peroxidation. The key contributing factors to ferroptosis are the loss of glutathione peroxidase 4 which is required for reversing lipid peroxidation, the buildup of redox-active iron and the oxidation of phospholipids containing polyunsaturated fatty acids. Ferroptosis has been associated with a number of diseases, including cancers such as hepatocellular carcinoma, breast cancer, acute renal damage and neurological disorders such as Alzheimer's disease and Alzheimer's disease, and there may be an association between ferroptosis and acute myeloid leukemia (AML). The present review aims to describe the primary regulatory pathways of ferroptosis, and the relationship between ferroptosis and the occurrence and development of AML. Furthermore, the present review comprehensively summarizes the latest advances in the treatment and prognosis of ferroptosis in AML.
Collapse
Affiliation(s)
- Wenlu Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Wen Wen
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Ran Tan
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Meirui Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Tantan Zhong
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Jianhong Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Haiping Chen
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
19
|
Zhang Y, Cao S, Zeng F, Pan D, Cai L, Zhou Y, Wang H, Qin G, Zhang C, Chen W. Dihydroartemisinin enhances the radiosensitivity of breast cancer by targeting ferroptosis signaling pathway through hsa_circ_0001610. Eur J Pharmacol 2024; 983:176943. [PMID: 39182549 DOI: 10.1016/j.ejphar.2024.176943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVE This study aimed to elucidate how DHA enhances the radiosensitivity of BC and to explain its potential mechanisms of action. METHODS The circular structure of hsa_circ_0001610 was confirmed by Sanger sequencing, RNase R treatment, RT-PCR analysis using gDNA or cDNA. Cellular localization of hsa_circ_0001610 and microRNA-139-5p (miR-139-5p) was detected by fluorescence in situ hybridization. Cell counting kit-8 assay, wound healing and colony formation tests for assessing cell proliferation, while flow cytometry was utilized to estimate cell cycle progression and apoptosis. Reactive oxygen species and malondialdehyde experiments were conducted to validate ferroptosis of BC cells. The expression of ncRNAs and mRNAs was quantified via qRT-PCR, and protein expression was analyzed using Western blot. The effects of hsa_circ_0001610 and DHA on radiosensitivity of BC in vivo were studied by establishing BC mice model. RESULTS In vivo and in vitro experimental results indicate that DHA promotes ferroptosis of BC cells at least partly by inhibiting hsa_circ_0001610/miR-139-5p/SLC7A11 pathway, thereby enhancing the radiosensitivity of BC cells. CONCLUSIONS Our findings showed that DHA can induce ferroptosis of BC cells by down-regulation of hsa_circ_0001610, thus enhancing radiosensitivity, suggesting a promising therapeutic strategy for enhancing BC radiosensitivity that is worthy of further exploration.
Collapse
Affiliation(s)
- YiWen Zhang
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - ShuYi Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, 510000, China
| | - FengXia Zeng
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - DeRun Pan
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - LongMei Cai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - YingYing Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - HongMei Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - GengGeng Qin
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China.
| | - Chao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, 510000, China.
| | - WeiGuo Chen
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
20
|
Kubota Y, Kimura S. Current Understanding of the Role of Autophagy in the Treatment of Myeloid Leukemia. Int J Mol Sci 2024; 25:12219. [PMID: 39596291 PMCID: PMC11594995 DOI: 10.3390/ijms252212219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The most important issues in acute myeloid leukemia are preventing relapse and treating relapse. Although the remission rate has improved to approximately 80%, the 5-year survival rate is only around 30%. The main reasons for this are the high relapse rate and the limited treatment options. In chronic myeloid leukemia patients, when a deep molecular response is achieved for a certain period of time through tyrosine kinase inhibitor treatment, about half of them will reach treatment-free remission, but relapse is still a problem. Therefore, potential therapeutic targets for myeloid leukemias are eagerly awaited. Autophagy suppresses the development of cancer by maintaining cellular homeostasis; however, it also promotes cancer progression by helping cancer cells survive under various metabolic stresses. In addition, autophagy is promoted or suppressed in cancer cells by various genetic mutations. Therefore, the development of therapies that target autophagy is also being actively researched in the field of leukemia. In this review, studies of the role of autophagy in hematopoiesis, leukemogenesis, and myeloid leukemias are presented, and the impact of autophagy regulation on leukemia treatment and the clinical trials of autophagy-related drugs to date is discussed.
Collapse
MESH Headings
- Humans
- Autophagy
- Animals
- Leukemia, Myeloid/pathology
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/therapy
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/drug therapy
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
- Hematopoiesis
Collapse
Affiliation(s)
- Yasushi Kubota
- Department of Clinical Laboratory Medicine, Saga-Ken Medical Centre Koseikan, Saga 840-8571, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan;
| |
Collapse
|
21
|
Ge T, Wang Y, Han Y, Bao X, Lu C. Exploring the Updated Roles of Ferroptosis in Liver Diseases: Mechanisms, Regulators, and Therapeutic Implications. Cell Biochem Biophys 2024:10.1007/s12013-024-01611-3. [PMID: 39543068 DOI: 10.1007/s12013-024-01611-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
Ferroptosis, a newly discovered mode of cell death, is a type of iron-dependent regulated cell death characterized by intracellular excessive lipid peroxidation and imbalanced redox. As the liver is susceptible to oxidative damage and the abnormal iron accumulation is a major feature of most liver diseases, studies on ferroptosis in the field of liver diseases are of great interest. Studies show that targeting the key regulators of ferroptosis can effectively alleviate or even reverse the deterioration process of liver diseases. System Xc- and glutathione peroxidase 4 are the main defense regulators of ferroptosis, while acyl-CoA synthetase long chain family member 4 is a key enzyme causing peroxidation in ferroptosis. Generally speaking, ferroptosis should be suppressed in alcoholic liver disease, non-alcoholic fatty liver disease, and drug-induced liver injury, while it should be induced in liver fibrosis and hepatocellular carcinoma. In this review, we summarize the main regulators involved in ferroptosis and then the mechanisms of ferroptosis in different liver diseases. Treatment options of drugs targeting ferroptosis are further concluded. Determining different triggers of ferroptosis can clarify the mechanism of ferroptosis occurs at both physiological and pathological levels.
Collapse
Affiliation(s)
- Ting Ge
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yang Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yiwen Han
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
22
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
23
|
Saedi S, Tan Y, Watson SE, Wintergerst KA, Cai L. Potential pathogenic roles of ferroptosis and cuproptosis in cadmium-induced or exacerbated cardiovascular complications in individuals with diabetes. Front Endocrinol (Lausanne) 2024; 15:1461171. [PMID: 39415790 PMCID: PMC11479913 DOI: 10.3389/fendo.2024.1461171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetes and its complications are major diseases that affect human health. Diabetic cardiovascular complications such as cardiovascular diseases (CVDs) are the major complications of diabetes, which are associated with the loss of cardiovascular cells. Pathogenically the role of ferroptosis, an iron-dependent cell death, and cuproptosis, a copper-dependent cell death has recently been receiving attention for the pathogenesis of diabetes and its cardiovascular complications. How exposure to environmental metals affects these two metal-dependent cell deaths in cardiovascular pathogenesis under diabetic and nondiabetic conditions remains largely unknown. As an omnipresent environmental metal, cadmium exposure can cause oxidative stress in the diabetic cardiomyocytes, leading to iron accumulation, glutathione depletion, lipid peroxidation, and finally exacerbate ferroptosis and disrupt the cardiac. Moreover, cadmium-induced hyperglycemia can enhance the circulation of advanced glycation end products (AGEs). Excessive AGEs in diabetes promote the upregulation of copper importer solute carrier family 31 member 1 through activating transcription factor 3/transcription factor PU.1, thereby increasing intracellular Cu+ accumulation in cardiomyocytes and disturbing Cu+ homeostasis, leading to a decline of Fe-S cluster protein and reactive oxygen species accumulation in cardiomyocytes mitochondria. In this review, we summarize the available evidence and the most recent advances exploring the underlying mechanisms of ferroptosis and cuproptosis in CVDs and diabetic cardiovascular complications, to provide critical perspectives on the potential pathogenic roles of ferroptosis and cuproptosis in cadmium-induced or exacerbated cardiovascular complications in diabetic individuals.
Collapse
Affiliation(s)
- Saman Saedi
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Wendy Novak Diabetes Institute, Norton Children’s Hospital, Louisville, KY, United States
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Sara E. Watson
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Wendy Novak Diabetes Institute, Norton Children’s Hospital, Louisville, KY, United States
- Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children’s Hospital, Louisville, KY, United States
| | - Kupper A. Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Wendy Novak Diabetes Institute, Norton Children’s Hospital, Louisville, KY, United States
- Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children’s Hospital, Louisville, KY, United States
- The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, United States
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Wendy Novak Diabetes Institute, Norton Children’s Hospital, Louisville, KY, United States
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States
- The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
24
|
Li P, Lyu T. Research Progress on Ferroptosis in Multiple Myeloma. Curr Treat Options Oncol 2024; 25:1276-1282. [PMID: 39287715 PMCID: PMC11485180 DOI: 10.1007/s11864-024-01250-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 09/19/2024]
Abstract
OPINION STATEMENT Multiple myeloma (MM) is the second most common hematological malignant (HM) tumor, and a large proportion of patients still suffer from treatment failure and a poor prognosis despite the use of some newly approved drugs, a deeper understanding of the underlying mechanism is still needed. Ferroptosis is a new form of programmed cell death (PCD) that is different from other traditional forms of cell death such as apoptosis, necrosis and autophagy. With the continuous deepening of research on ferroptosis, ferroptosis has been found to be closely related to MM. This article reviews the regulatory mechanism of ferroptosis and research progress on ferroptosis in MM, providing a new theoretical basis and strategies for the diagnosis and treatment of MM.
Collapse
Affiliation(s)
- Po Li
- Department of Orthopedic, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, Henan, China
| | - Tianxin Lyu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
25
|
Lan W, Yang L, Tan X. Crosstalk between ferroptosis and macrophages: potential value for targeted treatment in diseases. Mol Cell Biochem 2024; 479:2523-2543. [PMID: 37880443 DOI: 10.1007/s11010-023-04871-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023]
Abstract
Ferroptosis is a newly identified form of programmed cell death that is connected to iron-dependent lipid peroxidization. It involves a variety of physiological processes involving iron metabolism, lipid metabolism, oxidative stress, and biosynthesis of nicotinamide adenine dinucleotide phosphate, glutathione, and coenzyme Q10. So far, it has been discovered to contribute to the pathological process of many diseases, such as myocardial infarction, acute kidney injury, atherosclerosis, and so on. Macrophages are innate immune system cells that regulate metabolism, phagocytize pathogens and dead cells, mediate inflammatory reactions, promote tissue repair, etc. Emerging evidence shows strong associations between macrophages and ferroptosis, which can provide us with a deeper comprehension of the pathological process of diseases and new targets for the treatments. In this review, we summarized the crosstalk between macrophages and ferroptosis and anatomized the application of this association in disease treatments, both non-neoplastic and neoplastic diseases. In addition, we have also addressed problems that remain to be investigated, in the hope of inspiring novel therapeutic strategies for diseases.
Collapse
Affiliation(s)
- Wanxin Lan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Xuelian Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
26
|
Xia L, Qiu Y, Li J, Xu M, Dong Z. The Potential Role of Artemisinins Against Neurodegenerative Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1641-1660. [PMID: 39343990 DOI: 10.1142/s0192415x24500642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Artemisinin (ART) and its derivatives, collectively referred to as artemisinins (ARTs), have been approved for the treatment of malaria for decades. ARTs are converted into dihydroartemisinin (DHA), the only active form, which is reductive in vivo. In this review, we provide a brief overview of the neuroprotective potential of ARTs and the underlying mechanisms on several of the most common neurodegenerative diseases, particularly considering their potential application in those associated with cognitive and motor impairments including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). ARTs act as autophagy balancers to alleviate AD and PD. They inhibit neuroinflammatory responses by regulating phosphorylation of signal transduction proteins, such as AKT, PI3K, ERK, NF-κB, p38 MAPK, IκBα. In addition, ARTs regulate GABAergic signaling in a dose-dependent manner. Although they competitively inhibit the binding of gephyrin to GABAergic receptors, low doses of ARTs enhance GABAergic signaling. ARTs can also inhibit ferroptosis, activate the Akt/Bcl-2, AMPK, or ERK/CREB pathways to reduce oxidative stress, and maintain mitochondrial homeostasis, protecting neurons from oxidative stress injury. More importantly, ARTs structurally combine with and suppress β-Amyloid (A[Formula: see text]-induced neurotoxicity, reduce P-tau, and maintain O-GlcNAcylation/Phosphorylation balance, leading to relieved pathological changes in neurodegenerative diseases. Collectively, these natural properties endow ARTs with unique potential for application in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lei Xia
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education, Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
| | - Yiqiong Qiu
- Medical Laboratory of Changshou District Hospital of Traditional Chinese Medicine, Chongqing 401220, P. R. China
| | - Junjie Li
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education, Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
| | - Mingliang Xu
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education, Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
| | - Zhifang Dong
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education, Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
| |
Collapse
|
27
|
Gou X, Tang X, Liu C, Chen Z. Ferroptosis: a new mechanism of traditional Chinese medicine for treating hematologic malignancies. Front Oncol 2024; 14:1469178. [PMID: 39376985 PMCID: PMC11456518 DOI: 10.3389/fonc.2024.1469178] [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: 07/23/2024] [Accepted: 08/30/2024] [Indexed: 10/09/2024] Open
Abstract
Ferroptosis is a recently identified form of cell death characterized by lipid peroxidation and elevated iron levels. It is closely associated with hematologic malignancies, including leukemia, multiple myeloma (MM), and myelodysplastic syndromes (MDS). Research indicates that ferroptosis could represent a novel therapeutic target for these hematologic malignancies. Furthermore, traditional Chinese medicine (TCM) has been shown to modulate hematologic malignancies through the ferroptosis pathway. This paper aims to elucidate the mechanisms underlying ferroptosis and summarize the current research advancements regarding ferroptosis in hematologic malignancies, as well as the role of traditional Chinese medicine in the prevention and treatment of ferroptosis, with the goal of enhancing treatment efficacy.
Collapse
Affiliation(s)
- Xinyue Gou
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xudong Tang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chi Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhuo Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
28
|
Fatima S, Zhou H, Chen Y, Liu Q. Role of ferroptosis in the pathogenesis of heart disease. Front Physiol 2024; 15:1450656. [PMID: 39318361 PMCID: PMC11420141 DOI: 10.3389/fphys.2024.1450656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Ferroptosis is a new form of regulated necrosis characterized by iron-dependent lipid peroxidation, leading to irreparable lipid damage, membrane permeabilization, and necrotic cell death. Ferroptosis has recently been implicated in the pathogenesis of multiple forms of heart disease such as myocardial infarction, cardiac hypertrophy, heart failure, and various cardiomyopathies. Important progress has also been made regarding how ferroptosis is regulated in vitro and in vivo as well as its role in cardiac homeostasis and disease pathogenesis. In this review, we discuss molecular mechanisms that regulates ferroptosis in the heart, including pathways leading to iron overload and lipid peroxidation as well as the roles of key organelles in this process. We also discuss recent findings pertaining to the new pathogenic role of ferroptosis in various forms of heart disease as well as genetic and pharmacologic strategies targeting ferroptosis in the heart.
Collapse
Affiliation(s)
| | | | | | - Qinghang Liu
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| |
Collapse
|
29
|
Zhu L, Du Y. A promising new approach to cancer therapy: Manipulate ferroptosis by hijacking endogenous iron. Int J Pharm 2024; 662:124517. [PMID: 39084581 DOI: 10.1016/j.ijpharm.2024.124517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Ferroptosis, a form of regulated cell death characterized by iron-dependent phospholipid peroxidation, has emerged as a focal point in the field of cancer therapy. Compared with other cell death modes such as apoptosis and necrosis, ferroptosis exhibits many distinct characteristics in the molecular mechanisms and cell morphology, offering a promising avenue for combating cancers that are resistant to conventional therapeutic modalities. In light of the serious side effects associated with current Fenton-modulating ferroptosis therapies utilizing exogenous iron-based inorganic nanomaterials, hijacking endogenous iron could serve as an effective alternative strategy to trigger ferroptosis through targeting cellular iron regulatory mechanisms. A better understanding of the underlying iron regulatory mechanism in the process of ferroptosis has shed light on the current findings of endogenous ferroptosis-based nanomedicine strategies for cancer therapy. Here in this review article, we provide a comprehensive discussion on the regulatory network of iron metabolism and its pivotal role in ferroptosis, and present recent updates on the application of nanoparticles endowed with the ability to hijack endogenous iron for ferroptosis. We envision that the insights in the study may expedite the development and translation of endogenous ferroptosis-based nanomedicines for effective cancer treatment.
Collapse
Affiliation(s)
- Luwen Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang 321299, China.
| |
Collapse
|
30
|
Wang L, Tong L, Xiong Z, Chen Y, Zhang P, Gao Y, Liu J, Yang L, Huang C, Ye G, Du J, Liu H, Yang W, Wang Y. Ferroptosis-inducing nanomedicine and targeted short peptide for synergistic treatment of hepatocellular carcinoma. J Nanobiotechnology 2024; 22:533. [PMID: 39223666 PMCID: PMC11370132 DOI: 10.1186/s12951-024-02808-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The poor prognosis of hepatocellular carcinoma (HCC) is still an urgent challenge to be solved worldwide. Hence, assembling drugs and targeted short peptides together to construct a novel medicine delivery strategy is crucial for targeted and synergy therapy of HCC. Herein, a high-efficiency nanomedicine delivery strategy has been constructed by combining graphdiyne oxide (GDYO) as a drug-loaded platform, specific peptide (SP94-PEG) as a spear to target HCC cells, sorafenib, doxorubicin-Fe2+ (DOX-Fe2+), and siRNA (SLC7A11-i) as weapons to exert a three-path synergistic attack against HCC cells. In this work, SP94-PEG and GDYO form nanosheets with HCC-targeting properties, the chemotherapeutic drug DOX linked to ferrous ions increases the free iron pool in HCC cells and synergizes with sorafenib to induce cell ferroptosis. As a key gene of ferroptosis, interference with the expression of SLC7A11 makes the ferroptosis effect in HCC cells easier, stronger, and more durable. Through gene interference, drug synergy, and short peptide targeting, the toxic side effects of chemotherapy drugs are reduced. The multifunctional nanomedicine GDYO@SP94/DOX-Fe2+/sorafenib/SLC7A11-i (MNMG) possesses the advantages of strong targeting, good stability, the ability to continuously induce tumor cell ferroptosis and has potential clinical application value, which is different from traditional drugs.
Collapse
Affiliation(s)
- Luyang Wang
- Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
- Center for Drug Safety Evaluation, Qingdao Center for Pharmaceutical Collaborative Innovation, Qingdao, 266209, Shandong, P. R. China
- Department of Laboratory Medicine, Xinhua Hospital of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China
| | - Le Tong
- Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, P. R. China
| | - Zecheng Xiong
- CAS Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, CAS Research/ Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yi Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Ping Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Yan Gao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Jing Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Lei Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Chunqi Huang
- Department of Laboratory Medicine, Xinhua Hospital of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China
| | - Gaoqi Ye
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China.
| | - Huibiao Liu
- CAS Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, CAS Research/ Education Center for Excellence in Molecular Sciences, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China.
| | - Wei Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China.
| | - Ying Wang
- Department of Clinical Research Center, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China.
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, P. R. China.
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China.
| |
Collapse
|
31
|
Na X, Li L, Liu D, He J, Zhang L, Zhou Y. Natural products targeting ferroptosis pathways in cancer therapy (Review). Oncol Rep 2024; 52:123. [PMID: 39054952 PMCID: PMC11292301 DOI: 10.3892/or.2024.8782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Ferroptosis inducers (FIN) have a key role in cancer therapy and provide novel and innovative treatment strategies. Although many researchers have performed FIN screening of synthetic compounds, studies on the identification of FIN from natural products are limited, particularly in the field of drug development and combination therapy. In this review, this gap was addressed by comprehensively summarizing recent studies on ferroptosis. The causes of ferroptosis were categorized into driving and defensive factors, elucidating key pathways and targets. Next, through summarizing research on natural products that induce ferroptosis, the study elaborated in detail on the natural products that have FIN functions. Their discovery and development were also described and insight for clinical drug development was provided. In addition, the mechanisms of action were analyzed and potential combination therapies, resistance reversal and structural enhancements were presented. By highlighting the potential of natural products in inducing ferroptosis for cancer treatment, this review may serve as a reference for utilizing these compounds against cancer. It not only showed the significance of natural products but may also promote further investigation into their therapeutic effects, thus encouraging research in this field.
Collapse
Affiliation(s)
- Xin Na
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Lin Li
- Yunnan Cancer Hospital (Third Affiliated Hospital of Kunming Medical University), Kunming, Yunnan 650118, P.R. China
| | - Dongmei Liu
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Jiaqi He
- The First Clinical Medical College of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Ling Zhang
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yiping Zhou
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
32
|
Guo J, Huang M, Hou S, Yuan J, Chang X, Gao S, Zhang Z, Wu Z, Li J. Therapeutic Potential of Terpenoids in Cancer Treatment: Targeting Mitochondrial Pathways. Cancer Rep (Hoboken) 2024; 7:e70006. [PMID: 39234662 PMCID: PMC11375335 DOI: 10.1002/cnr2.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/30/2024] [Accepted: 08/11/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND In recent decades, natural compounds have been considered a significant source of new antitumor medicines due to their unique advantages. Several in vitro and in vivo studies have focused on the effect of terpenoids on apoptosis mediated by mitochondria in malignant cells. RECENT FINDINGS In this review article, we focused on six extensively studied terpenoids, including sesquiterpenes (dihydroartemisinin and parthenolide), diterpenes (oridonin and triptolide), and triterpenes (betulinic acid and oleanolic acid), and their efficacy in targeting mitochondria to induce cell death. Terpenoid-induced mitochondria-related cell death includes apoptosis, pyroptosis, necroptosis, ferroptosis, autophagy, and necrosis caused by mitochondrial permeability transition. Apoptosis and autophagy interact in meaningful ways. In addition, in view of several disadvantages of terpenoids, such as low stability and bioavailability, advances in research on combination chemotherapy and chemical modification were surveyed. CONCLUSION This article deepens our understanding of the association between terpenoids and mitochondrial cell death, presenting a hypothetical basis for the use of terpenoids in anticancer management.
Collapse
Affiliation(s)
- Jianxin Guo
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Ming Huang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Shuang Hou
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Jianfeng Yuan
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Xiaoyue Chang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Shuang Gao
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Zhenhan Zhang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Zhongbing Wu
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Jing Li
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
33
|
Li H, Li Y, Yu Y, Ren X, Yang C, Jin W, Li K, Zhou Y, Wu C, Shen Y, Hu W, Liu Y, Yu L, Tong X, Du J, Wang Y. GSH exhaustion via inhibition of xCT-GSH-GPX4 pathway synergistically enhanced DSF/Cu-induced cuproptosis in myelodysplastic syndromes. Free Radic Biol Med 2024; 222:130-148. [PMID: 38866192 DOI: 10.1016/j.freeradbiomed.2024.06.006] [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: 04/23/2024] [Revised: 06/02/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024]
Abstract
The clinical application of the therapeutic approach in myelodysplastic syndromes (MDS) remains an insurmountable challenge for the high propensity for progressing to acute myeloid leukemia and predominantly affecting elderly individuals. Thus, the discovery of molecular mechanisms underlying the regulatory network of different programmed cell death holds great promise for the identification of therapeutic targets and provides insights into new therapeutic avenues. Herein, we found that disulfiram/copper (DSF/Cu) significantly repressed the cell viability, increased reactive oxygen species (ROS) accumulation, destroyed mitochondrial morphology, and altered oxygen consumption rate. Further studies verified that DSF/Cu induces cuproptosis, as evidenced by the depletion of glutathione (GSH), aggregation of lipoylated DLAT, and induced loss of Fe-S cluster-containing proteins, which could be rescued by tetrathiomolybdate and knockdown of ferredoxin 1 (FDX1). Additionally, GSH contributed to the tolerance of DSF/Cu-mediated cuproptosis, while pharmacological chelation of GSH triggered ROS accumulation and sensitized cell death. The xCT-GSH-GPX4 axis is the ideal downstream component of ferroptosis that exerts a powerful protective mechanism. Notably, classical xCT inhibitors were capable of leading to the catastrophic accumulation of ROS and exerting synergistic cell death, while xCT overexpression restored these phenomena. Simvastatin, an inhibitor of HMG-CoA (3-hydroxy-3-methylglutaryl-coenzyme A) reductase, has beneficial effects in repurposing for inhibiting GPX4. Similarly, the combination treatment of DSF/Cu and simvastatin dramatically decreased the expression of GPX4 and Fe-S proteins, ultimately accelerating cell death. Moreover, we identified that the combination treatment of DSF/Cu and simvastatin also had a synergistic antitumor effect in the MDS mouse model, with the reduced GPX4, increased COX-2 and accumulated lipid peroxides. Overall, our study provided insight into developing a novel synergistic strategy to sensitize MDS therapy by targeting ferroptosis and cuproptosis.
Collapse
Affiliation(s)
- Huanjuan Li
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yanchun Li
- Department of Clinical Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, China
| | - Yanhua Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xueying Ren
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, China
| | - Chen Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Weidong Jin
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Keyi Li
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yi Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Cuiyun Wu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhuan Shen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wanye Hu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
| | - Yingchao Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Lingyan Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiangmin Tong
- Department of Clinical Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Ying Wang
- Department of Clinical Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
34
|
Khan S, Bano N, Ahamad S, John U, Dar NJ, Bhat SA. Excitotoxicity, Oxytosis/Ferroptosis, and Neurodegeneration: Emerging Insights into Mitochondrial Mechanisms. Aging Dis 2024:AD.2024.0125-1. [PMID: 39122453 DOI: 10.14336/ad.2024.0125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Mitochondrial dysfunction plays a pivotal role in the development of age-related diseases, particularly neurodegenerative disorders. The etiology of mitochondrial dysfunction involves a multitude of factors that remain elusive. This review centers on elucidating the role(s) of excitotoxicity, oxytosis/ferroptosis and neurodegeneration within the context of mitochondrial bioenergetics, biogenesis, mitophagy and oxidative stress and explores their intricate interplay in the pathogenesis of neurodegenerative diseases. The effective coordination of mitochondrial turnover processes, notably mitophagy and biogenesis, is assumed to be critically important for cellular resilience and longevity. However, the age-associated decrease in mitophagy impedes the elimination of dysfunctional mitochondria, consequently impairing mitochondrial biogenesis. This deleterious cascade results in the accumulation of damaged mitochondria and deterioration of cellular functions. Both excitotoxicity and oxytosis/ferroptosis have been demonstrated to contribute significantly to the pathophysiology of neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's Disease (HD), Amyotrophic Lateral Sclerosis (ALS) and Multiple Sclerosis (MS). Excitotoxicity, characterized by excessive glutamate signaling, initiates a cascade of events involving calcium dysregulation, energy depletion, and oxidative stress and is intricately linked to mitochondrial dysfunction. Furthermore, emerging concepts surrounding oxytosis/ferroptosis underscore the importance of iron-dependent lipid peroxidation and mitochondrial engagement in the pathogenesis of neurodegeneration. This review not only discusses the individual contributions of excitotoxicity and ferroptosis but also emphasizes their convergence with mitochondrial dysfunction, a key driver of neurodegenerative diseases. Understanding the intricate crosstalk between excitotoxicity, oxytosis/ferroptosis, and mitochondrial dysfunction holds potential to pave the way for mitochondrion-targeted therapeutic strategies. Such strategies, with a focus on bioenergetics, biogenesis, mitophagy, and oxidative stress, emerge as promising avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh-202002, India
| | - Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh-202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh-202002, India
| | - Urmilla John
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India; School of Studies in Zoology, Jiwaji University, Gwalior, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | | |
Collapse
|
35
|
Guan S, Zhang S, Liu M, Guo J, Chen Y, Shen X, Deng X, Lu J. Preventive effects of lactoferrin on acute alcohol-induced liver injury via iron chelation and regulation of iron metabolism. J Dairy Sci 2024; 107:5316-5329. [PMID: 38608952 DOI: 10.3168/jds.2023-24490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/02/2024] [Indexed: 04/14/2024]
Abstract
Lactoferrin is widely found in milk and has the ability to bind iron. Previous studies have reported that lactoferrin was effective in the prevention and treatment of acute alcohol-induced liver injury (AALI). Ferroptosis is a recently discovered cell death and is involved in the development of AALI. However, the potential role of lactoferrin in acute alcohol-induced ferroptosis is still unclear. In this study, we observed that lactoferrin (10, 20, and 40 μg/mL) significantly mitigated alcohol (300 mM)-induced injury in vitro. Additionally, lactoferrin (100 and 200 mg/kg BW) significantly alleviated alcohol (4.8 g/kg BW)-induced injury in vivo. Our results showed that lactoferrin inhibited alcohol-induced upregulation of the ferroptosis marker protein ACSL4 and downregulation of GPX4. Meanwhile, lactoferrin treatment successfully reversed the elevated malondialdehyde (MDA) levels and the reduced glutathione (GSH) levels caused by alcohol treatment. These results may indicate that lactoferrin significantly decreased ferroptosis in vivo and in vitro. Lactoferrin has the potential to chelate iron, and our results showed that lactoferrin (20 μg/mL) significantly reduced iron ions and the expression of the ferritin heavy chain (FTH) under FeCl3 (100 μM) treatment. It was demonstrated that lactoferrin had a significant iron-chelating effect and reduced iron overload caused by FeCl3 in AML12 cells. Next, we examined iron content and the expression of iron metabolism marker proteins transferrin receptor (TFR), divalent metal transporter 1 (DMT1), FTH, and ferroportin (FPN). Our results showed that lactoferrin alleviated iron overload induced by acute alcohol. The expression of TFR and DMT1 was downregulated, and FPN and FTH were upregulated after lactoferrin treatment in vivo and in vitro. Above all, the study suggested that lactoferrin can alleviate AALI by mitigating acute alcohol-induced ferroptosis. Lactoferrin may offer new strategies for the prevention or treatment of AALI.
Collapse
Affiliation(s)
- Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China; State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Shengzhuo Zhang
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Meitong Liu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Jiakang Guo
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Yuelin Chen
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Xue Shen
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China.
| | - Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, China.
| |
Collapse
|
36
|
Lu CL, Sha JJ, Ma RF, Dong XT, Su XR, Cong B, Wang SJ. Severe Hypothermia Induces Ferroptosis in Cerebral Cortical Nerve Cells. Int J Mol Sci 2024; 25:8086. [PMID: 39125656 PMCID: PMC11311695 DOI: 10.3390/ijms25158086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Abnormal shifts in global climate, leading to extreme weather, significantly threaten the safety of individuals involved in outdoor activities. Hypothermia-induced coma or death frequently occurs in clinical and forensic settings. Despite this, the precise mechanism of central nervous system injury due to hypothermia remains unclear, hindering the development of targeted clinical treatments and specific forensic diagnostic indicators. The GEO database was searched to identify datasets related to hypothermia. Post-bioinformatics analyses, DEGs, and ferroptosis-related DEGs (FerrDEGs) were intersected. GSEA was then conducted to elucidate the functions of the Ferr-related genes. Animal experiments conducted in this study demonstrated that hypothermia, compared to the control treatment, can induce significant alterations in iron death-related genes such as PPARG, SCD, ADIPOQ, SAT1, EGR1, and HMOX1 in cerebral cortex nerve cells. These changes lead to iron ion accumulation, lipid peroxidation, and marked expression of iron death-related proteins. The application of the iron death inhibitor Ferrostatin-1 (Fer-1) effectively modulates the expression of these genes, reduces lipid peroxidation, and improves the expression of iron death-related proteins. Severe hypothermia disrupts the metabolism of cerebral cortex nerve cells, causing significant alterations in ferroptosis-related genes. These genetic changes promote ferroptosis through multiple pathways.
Collapse
Affiliation(s)
| | | | | | | | | | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, College of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Road, Shijiazhuang 050017, China; (C.-L.L.); (J.-J.S.); (R.-F.M.); (X.-T.D.); (X.-R.S.)
| | - Song-Jun Wang
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, College of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Road, Shijiazhuang 050017, China; (C.-L.L.); (J.-J.S.); (R.-F.M.); (X.-T.D.); (X.-R.S.)
| |
Collapse
|
37
|
Tan X, He Y, Yu P, Deng Y, Xie Z, Guo J, Hou Q, Li P, Lin X, Ouyang S, Ma W, Xie Y, Guo Z, Chen D, Zhang Z, Zhu Y, Huang F, Zhao Z, Zhang C, Guo Z, Chen X, Peng T, Li L, Xie W. The dual role of FSP1 in programmed cell death: resisting ferroptosis in the cell membrane and promoting necroptosis in the nucleus of THP-1 cells. Mol Med 2024; 30:102. [PMID: 39009982 PMCID: PMC11247902 DOI: 10.1186/s10020-024-00861-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/10/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Acute monocytic leukemia-M5 (AML-M5) remains a challenging disease due to its high morbidity and poor prognosis. In addition to the evidence mentioned earlier, several studies have shown that programmed cell death (PCD) serves a critical function in treatment of AML-M5. However, the role and relationship between ferroptosis and necroptosis in AML-M5 remains unclear. METHODS THP-1 cells were mainly treated with Erastin and IMP-366. The changes of ferroptosis and necroptosis levels were detected by CCK-8, western blot, quantitative real-time PCR, and electron microscopy. Flow cytometry was applied to detect the ROS and lipid ROS levels. MDA, 4-HNE, GSH and GSSG were assessed by ELISA kits. Intracellular distribution of FSP1 was studied by immunofluorescent staining and western blot. RESULTS The addition of the myristoylation inhibitor IMP-366 to erastin-treated acute monocytic leukemia cell line THP-1 cell not only resulted in greater susceptibility to ferroptosis characterized by lipid peroxidation, glutathione (GSH) depletion and mitochondrial shrinkage, as the FSP1 position on membrane was inhibited, but also increased p-RIPK1 and p-MLKL protein expression, as well as a decrease in caspase-8 expression, and triggered the characteristic necroptosis phenomena, including cytoplasmic translucency, mitochondrial swelling, membranous fractures by FSP1 migration into the nucleus via binding importin α2. It is interesting to note that ferroptosis inhibitor fer-1 reversed necroptosis. CONCLUSION We demonstrated that inhibition of myristoylation by IMP-366 is capable of switching ferroptosis and ferroptosis-dependent necroptosis in THP-1 cells. In these findings, FSP1-mediated ferroptosis and necroptosis are described as alternative mechanisms of PCD of THP-1 cells, providing potential therapeutic strategies and targets for AML-M5.
Collapse
Affiliation(s)
- Xiaoqian Tan
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan, China
| | - Yinling He
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Panpan Yu
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yunong Deng
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhongcheng Xie
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiami Guo
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Qin Hou
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pin Li
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyan Lin
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Ouyang
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wentao Ma
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yushu Xie
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Zilong Guo
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Dandan Chen
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Zhixia Zhang
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Yunyu Zhu
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Fei Huang
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Ziye Zhao
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Cen Zhang
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Zhirong Guo
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Xi Chen
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tianhong Peng
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Liang Li
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wei Xie
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
38
|
Fujii J, Imai H. Oxidative Metabolism as a Cause of Lipid Peroxidation in the Execution of Ferroptosis. Int J Mol Sci 2024; 25:7544. [PMID: 39062787 PMCID: PMC11276677 DOI: 10.3390/ijms25147544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Ferroptosis is a type of nonapoptotic cell death that is characteristically caused by phospholipid peroxidation promoted by radical reactions involving iron. Researchers have identified many of the protein factors that are encoded by genes that promote ferroptosis. Glutathione peroxidase 4 (GPX4) is a key enzyme that protects phospholipids from peroxidation and suppresses ferroptosis in a glutathione-dependent manner. Thus, the dysregulation of genes involved in cysteine and/or glutathione metabolism is closely associated with ferroptosis. From the perspective of cell dynamics, actively proliferating cells are more prone to ferroptosis than quiescent cells, which suggests that radical species generated during oxygen-involved metabolism are responsible for lipid peroxidation. Herein, we discuss the initial events involved in ferroptosis that dominantly occur in the process of energy metabolism, in association with cysteine deficiency. Accordingly, dysregulation of the tricarboxylic acid cycle coupled with the respiratory chain in mitochondria are the main subjects here, and this suggests that mitochondria are the likely source of both radical electrons and free iron. Since not only carbohydrates, but also amino acids, especially glutamate, are major substrates for central metabolism, dealing with nitrogen derived from amino groups also contributes to lipid peroxidation and is a subject of this discussion.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Hirotaka Imai
- Laboratory of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, Tokyo 108-8641, Japan
- Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo 108-8641, Japan
| |
Collapse
|
39
|
Zhong F, Zhang X, Wang Z, Li X, Huang B, Kong G, Wang X. The therapeutic and biomarker significance of ferroptosis in chronic myeloid leukemia. Front Immunol 2024; 15:1402669. [PMID: 39026664 PMCID: PMC11254662 DOI: 10.3389/fimmu.2024.1402669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Background The relationship between ferroptosis and the progression and treatment of hematological tumors has been extensively studied, although its precise association with chronic myeloid leukemia (CML) remains uncertain. Methods Multi-transcriptome sequencing data were utilized to analyze the ferroptosis level of CML samples and its correlation with the tumor microenvironment, disease progression, and treatment response. Machine learning algorithms were employed to identify diagnostic ferroptosis-related genes (FRGs). The consensus clustering algorithm was applied to identify ferroptosis-related molecular subtypes. Clinical samples were collected for sequencing to validate the results obtained from bioinformatics analysis. Cell experiments were conducted to investigate the therapeutic efficacy of induced ferroptosis in drug-resistant CML. Results Ferroptosis scores were significantly lower in samples from patients with CML compared to normal samples, and these scores further decreased with disease progression and non-response to treatment. Most FRGs were downregulated in CML samples. A high ferroptosis score was also associated with greater immunosuppression and increased activity of metabolic pathways. Through support vector machine recursive feature elimination (SVM-RFE), least absolute shrinkage selection operator (LASSO), and random forest (RF) algorithms, we identified five FRGs (ACSL6, SLC11A2, HMOX1, SLC38A1, AKR1C3) that have high diagnostic value. The clinical diagnostic value of these five FRGs and their effectiveness in differentiating CML from other hematological malignancies were validated using additional validation cohorts and our real-world cohort. There are significant differences in immune landscape, chemosensitivity, and immunotherapy responsiveness between the two ferroptosis-related molecular subtypes. By conducting cellular experiments, we confirmed that CML-resistant cells are more sensitive to induction of ferroptosis and can enhance the sensitivity of imatinib treatment. Conclusion Our study unveils the molecular signature of ferroptosis in samples from patients with CML. FRG identified by a variety of machine learning algorithms has reliable clinical diagnostic value. Furthermore, the characterization of different ferroptosis-related molecular subtypes provides valuable insights into individual patient characteristics and can guide clinical treatment strategies. Targeting and inducing ferroptosis holds great promise as a therapeutic approach for drug-resistant CML.
Collapse
MESH Headings
- Ferroptosis/genetics
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Biomarkers, Tumor
- Tumor Microenvironment
- Drug Resistance, Neoplasm/genetics
- Computational Biology/methods
- Machine Learning
Collapse
Affiliation(s)
- Fangmin Zhong
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xueru Zhang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zihao Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaolin Li
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Guangyao Kong
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
40
|
Tang Y, Zhuang Y, Zhao C, Gu S, Zhang J, Bi S, Wang M, Bao L, Li M, Zhang W, Zhu L. The metabolites from traditional Chinese medicine targeting ferroptosis for cancer therapy. Front Pharmacol 2024; 15:1280779. [PMID: 39021832 PMCID: PMC11251977 DOI: 10.3389/fphar.2024.1280779] [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: 08/21/2023] [Accepted: 05/15/2024] [Indexed: 07/20/2024] Open
Abstract
Cancer is a major disease with ever-increasing morbidity and mortality. The metabolites derived from traditional Chinese medicine (TCM) have played a significant role in combating cancers with curative efficacy and unique advantages. Ferroptosis, an iron-dependent programmed death characterized by the accumulation of lipid peroxide, stands out from the conventional forms of cell death, such as apoptosis, pyroptosis, necrosis, and autophagy. Recent evidence has demonstrated the potential of TCM metabolites targeting ferroptosis for cancer therapy. We collected and screened related articles published in or before June 2023 using PubMed, Google Scholar, and Web of Science. The searched keywords in scientific databases were ferroptosis, cancer, tumor, traditional Chinese medicine, botanical drugs, and phytomedicine. Only research related to ferroptosis, the metabolites from TCM, and cancer was considered. In this review, we introduce an overview of the current knowledge regarding the ferroptosis mechanisms and review the research advances on the metabolites of TCM inhibiting cancer by targeting ferroptosis.
Collapse
Affiliation(s)
- Yu Tang
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ying Zhuang
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chuanxiang Zhao
- Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai’an, Jiangsu, China
| | - Shuangshuang Gu
- Shanghai Institute of Rheumatology, Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Junya Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shiqi Bi
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ming Wang
- Department of Medical Imaging, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Lei Bao
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mei Li
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Liqun Zhu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
41
|
Le Y, Liu Q, Yang Y, Wu J. The emerging role of nuclear receptor coactivator 4 in health and disease: a novel bridge between iron metabolism and immunity. Cell Death Discov 2024; 10:312. [PMID: 38961066 PMCID: PMC11222541 DOI: 10.1038/s41420-024-02075-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
Nuclear receptor coactivator 4 (NCOA4) has recently been recognized as a selective cargo receptor of ferritinophagy participating in ferroptosis. However, NCOA4 is also a coactivator that modulates the transcriptional activity of many vital nuclear receptors. Recent novel studies have documented the role of NCOA4 in healthy and pathogenic conditions via its modulation of iron- and non-iron-dependent metabolic pathways. NCOA4 exhibits non-ferritinophagic and iron-independent features such as promoting tumorigenesis and erythropoiesis, immunomodulation, regulating autophagy, and participating in DNA replication and mitosis. Full-length human-NCOA4 is composed of 614 amino acids, of which the N-terminal (1-237) contains nuclear-receptor-binding domains, while the C-terminal (238-614) principally contains a ferritin-binding domain. The exploration of the protein structure of NCOA4 suggests that NCOA4 possesses additional significant and complex functions based on its structural domains. Intriguingly, another three isoforms of NCOA4 that are produced by alternative splicing have been identified, which may also display disparate activities in physiological and pathological processes. Thus, NCOA4 has become an important bridge that encompasses interactions between immunity and metabolism. In this review, we outline the latest advances in the important regulating mechanisms underlying NCOA4 actions in health and disease conditions, providing insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Yue Le
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Qinjie Liu
- Department of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yi Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Jie Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
- Research Center of Surgery, BenQ Medical Center, the Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210021, China.
| |
Collapse
|
42
|
Tang N, Liu X, Liu Y, Wang H, Zhao Y, Wang H, Hu Z. Dihydroartemisinin induces ferroptosis in T cell acute lymphoblastic leukemia cells by downregulating SLC7A11 and activating the ATF4‑CHOP signaling pathway. Oncol Lett 2024; 28:337. [PMID: 38846431 PMCID: PMC11153983 DOI: 10.3892/ol.2024.14470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/02/2024] [Indexed: 06/09/2024] Open
Abstract
The present study aimed to investigate the anti-leukemic effects of dihydroartemisinin (DHA) on T-cell acute lymphoblastic leukemia (T-ALL) cell lines, Jurkat and Molt-4, and the underlying mechanisms. Cell Counting Kit-8 was performed to measure cell viability. Cell apoptosis and cell cycle distribution were assessed by flow cytometry. The expression levels of ATF4 and CHOP mRNA were assessed by reverse transcription-quantitative PCR, while the protein abundance of SLC7A11, GPX4, ATF4 and CHOP was determined by western blotting. Moreover, malondialdehyde, glutathione (GSH) and reactive oxygen species (ROS) assays were used to detect the levels of ferroptosis. The results showed that DHA suppressed T-ALL cell viability in vitro, and induced cell cycle arrest at S or G2/M phase. DHA also induced ROS burst, activated endoplasmic reticulum (ER) stress, disrupted the system Xc--GSH-GSH peroxidase 4 antioxidant system, and increased lipid peroxide accumulation, resulting in cell death. By contrast, the pharmacological inhibition of ferroptosis alleviated DHA-induced cell death, confirming that DHA induces T-ALL cell death via ferroptosis. Mechanistically, the effect of DHA on ferroptosis was partly mediated by downregulating SLC7A11 and upregulating the ATF4-CHOP signaling pathway, which is associated with ER stress. These results indicated that DHA may induce ferroptosis in T-ALL cell lines and could represent a promising therapeutic agent for treating T-ALL.
Collapse
Affiliation(s)
- Na Tang
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261042, P.R. China
- Graduate School, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Xinling Liu
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261042, P.R. China
| | - Yong Liu
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261042, P.R. China
| | - Haihua Wang
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261042, P.R. China
| | - Yao Zhao
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261042, P.R. China
| | - Haiying Wang
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261042, P.R. China
| | - Zhenbo Hu
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261042, P.R. China
| |
Collapse
|
43
|
Li J, Feng Y, Li Y, He P, Zhou Q, Tian Y, Yao R, Yao Y. Ferritinophagy: A novel insight into the double-edged sword in ferritinophagy-ferroptosis axis and human diseases. Cell Prolif 2024; 57:e13621. [PMID: 38389491 PMCID: PMC11216947 DOI: 10.1111/cpr.13621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/19/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
Nuclear receptor coactive 4 (NCOA4), which functions as a selective cargo receptor, is a critical regulator of the particularly autophagic degradation of ferritin, a process known as ferritinophagy. Mechanistically, NCOA4-mediated ferritinophagy performs an increasingly vital role in the maintenance of intracellular iron homeostasis by promoting ferritin transport and iron release as needed. Ferritinophagy is not only involved in iron-dependent responses but also in the pathogenesis and progression of various human diseases, including metabolism-related, neurodegenerative, cardiovascular and infectious diseases. Therefore, ferritinophagy is of great importance in maintaining cell viability and function and represents a potential therapeutic target. Recent studies indicated that ferritinophagy regulates the signalling pathway associated with ferroptosis, a newly discovered type of cell death characterised by iron-dependent lipid peroxidation. Although accumulating evidence clearly demonstrates the importance of the interplay between dysfunction in iron metabolism and ferroptosis, a deeper understanding of the double-edged sword effect of ferritinophagy in ferroptosis has remained elusive. Details of the mechanisms underlying the ferritinophagy-ferroptosis axis in regulating relevant human diseases remain to be elucidated. In this review, we discuss the latest research findings regarding the mechanisms that regulate the biological function of NCOA4-mediated ferritinophagy and its contribution to the pathophysiology of ferroptosis. The important role of the ferritinophagy-ferroptosis axis in human diseases will be discussed in detail, highlighting the great potential of targeting ferritinophagy in the treatment of diseases.
Collapse
Affiliation(s)
- Jing‐Yan Li
- Department of EmergencyThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yan‐Hua Feng
- Department of OrthopedicsHebei Provincial Chidren's HospitalShijiazhuangChina
| | - Yu‐Xuan Li
- Translational Medicine Research CenterMedical Innovation Research Division and Fourth Medical Center of the Chinese PLA General HospitalBeijingChina
| | - Peng‐Yi He
- Translational Medicine Research CenterMedical Innovation Research Division and Fourth Medical Center of the Chinese PLA General HospitalBeijingChina
| | - Qi‐Yuan Zhou
- Department of EmergencyThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Ying‐Ping Tian
- Department of EmergencyThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Ren‐Qi Yao
- Translational Medicine Research CenterMedical Innovation Research Division and Fourth Medical Center of the Chinese PLA General HospitalBeijingChina
| | - Yong‐Ming Yao
- Department of EmergencyThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
- Translational Medicine Research CenterMedical Innovation Research Division and Fourth Medical Center of the Chinese PLA General HospitalBeijingChina
| |
Collapse
|
44
|
Feng Z, Luan M, Zhu W, Xing Y, Ma X, Wang Y, Jia Y. Targeted ferritinophagy in gastrointestinal cancer: from molecular mechanisms to implications. Arch Toxicol 2024; 98:2007-2018. [PMID: 38602537 DOI: 10.1007/s00204-024-03745-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Gastrointestinal cancer is a significant global health burden, necessitating the development of novel therapeutic strategies. Emerging evidence has highlighted the potential of targeting ferritinophagy as a promising approach for the treatment of gastrointestinal cancer. Ferritinophagy is a form of selective autophagy that is mediated by the nuclear receptor coactivator 4 (NCOA4). This process plays a crucial role in regulating cellular iron homeostasis and has been implicated in various pathological conditions, including cancer. This review discusses the molecular mechanisms underlying ferritinophagy and its relevance to gastrointestinal cancer. Furthermore, we highlight the potential therapeutic implications of targeting ferritinophagy in gastrointestinal cancer. Several approaches have been proposed to modulate ferritinophagy, including small molecule inhibitors and immunotherapeutic strategies. We discuss the advantages and challenges associated with these therapeutic interventions and provide insights into their potential clinical applications.
Collapse
Affiliation(s)
- Zhaotian Feng
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Muhua Luan
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Wenshuai Zhu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Yunshan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Yanfei Jia
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China.
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China.
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China.
| |
Collapse
|
45
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Decoding ferroptosis: Revealing the hidden assassin behind cardiovascular diseases. Biomed Pharmacother 2024; 176:116761. [PMID: 38788596 DOI: 10.1016/j.biopha.2024.116761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
The discovery of regulatory cell death processes has driven innovation in cardiovascular disease (CVD) therapeutic strategies. Over the past decade, ferroptosis, an iron-dependent form of regulated cell death driven by excessive lipid peroxidation, has been shown to drive the development of multiple CVDs. This review provides insights into the evolution of the concept of ferroptosis, the similarities and differences with traditional modes of programmed cell death (e.g., apoptosis, autophagy, and necrosis), as well as the core regulatory mechanisms of ferroptosis (including cystine/glutamate transporter blockade, imbalance of iron metabolism, and lipid peroxidation). In addition, it provides not only a detailed review of the role of ferroptosis and its therapeutic potential in widely studied CVDs such as coronary atherosclerotic heart disease, myocardial infarction, myocardial ischemia/reperfusion injury, heart failure, cardiomyopathy, and aortic aneurysm but also an overview of the phenomenon and therapeutic perspectives of ferroptosis in lesser-addressed CVDs such as cardiac valvulopathy, pulmonary hypertension, and sickle cell disease. This article aims to integrate this knowledge to provide a comprehensive view of ferroptosis in a wide range of CVDs and to drive innovation and progress in therapeutic strategies in this field.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
46
|
Liu D, Hu Z, Lu J, Yi C. Redox-Regulated Iron Metabolism and Ferroptosis in Ovarian Cancer: Molecular Insights and Therapeutic Opportunities. Antioxidants (Basel) 2024; 13:791. [PMID: 39061859 PMCID: PMC11274267 DOI: 10.3390/antiox13070791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Ovarian cancer (OC), known for its lethality and resistance to chemotherapy, is closely associated with iron metabolism and ferroptosis-an iron-dependent cell death process, distinct from both autophagy and apoptosis. Emerging evidence suggests that dysregulation of iron metabolism could play a crucial role in OC by inducing an imbalance in the redox system, which leads to ferroptosis, offering a novel therapeutic approach. This review examines how disruptions in iron metabolism, which affect redox balance, impact OC progression, focusing on its essential cellular functions and potential as a therapeutic target. It highlights the molecular interplay, including the role of non-coding RNAs (ncRNAs), between iron metabolism and ferroptosis, and explores their interactions with key immune cells such as macrophages and T cells, as well as inflammation within the tumor microenvironment. The review also discusses how glycolysis-related iron metabolism influences ferroptosis via reactive oxygen species. Targeting these pathways, especially through agents that modulate iron metabolism and ferroptosis, presents promising therapeutic prospects. The review emphasizes the need for deeper insights into iron metabolism and ferroptosis within the redox-regulated system to enhance OC therapy and advocates for continued research into these mechanisms as potential strategies to combat OC.
Collapse
Affiliation(s)
- Dan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China; (D.L.); (Z.H.)
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
| | - Zewen Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China; (D.L.); (Z.H.)
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
| | - Jinzhi Lu
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
- Department of Laboratory Medicine, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China
| | - Cunjian Yi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China; (D.L.); (Z.H.)
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
| |
Collapse
|
47
|
Yang Y, Lin Y, Han Z, Wang B, Zheng W, Wei L. Ferroptosis: a novel mechanism of cell death in ophthalmic conditions. Front Immunol 2024; 15:1440309. [PMID: 38994366 PMCID: PMC11236620 DOI: 10.3389/fimmu.2024.1440309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
Ferroptosis, a new type of programmed cell death proposed in recent years, is characterized mainly by reactive oxygen species and iron-mediated lipid peroxidation and differs from programmed cell death, such as apoptosis, necrosis, and autophagy. Ferroptosis is associated with a variety of physiological and pathophysiological processes. Recent studies have shown that ferroptosis can aggravate or reduce the occurrence and development of diseases by targeting metabolic pathways and signaling pathways in tumors, ischemic organ damage, and other degenerative diseases related to lipid peroxidation. Increasing evidence suggests that ferroptosis is closely linked to the onset and progression of various ophthalmic conditions, including corneal injury, glaucoma, age-related macular degeneration, diabetic retinopathy, retinal detachment, and retinoblastoma. Our review of the current research on ferroptosis in ophthalmic diseases reveals significant advancements in our understanding of the pathogenesis, aetiology, and treatment of these conditions.
Collapse
Affiliation(s)
- Yaqi Yang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yumeng Lin
- Naniing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
- Naniing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Bo Wang
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Wei Zheng
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Lijuan Wei
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
48
|
Shah SS, Stone EF, Francis RO, Karafin MS. The global role of G6PD in infection and immunity. Front Immunol 2024; 15:1393213. [PMID: 38938571 PMCID: PMC11208698 DOI: 10.3389/fimmu.2024.1393213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) deficiency is the most common enzymopathy in humans. G6PD is an essential enzyme in the pentose phosphate pathway (PPP), generating NADPH needed for cellular biosynthesis and reactive oxygen species (ROS) homeostasis, the latter especially key in red blood cells (RBCs). Beyond the RBC, there is emerging evidence that G6PD exerts an immunologic role by virtue of its functions in leukocyte oxidative metabolism and anabolic synthesis necessary for immune effector function. We review these here, and consider the global immunometabolic role of G6PD activity and G6PD deficiency in modulating inflammation and immunopathology.
Collapse
Affiliation(s)
- Shivang S. Shah
- Department of Pediatrics, Columbia University, New York, NY, United States
| | - Elizabeth F. Stone
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Richard O. Francis
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Matthew S. Karafin
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
49
|
Chen X, Tsvetkov AS, Shen HM, Isidoro C, Ktistakis NT, Linkermann A, Koopman WJ, Simon HU, Galluzzi L, Luo S, Xu D, Gu W, Peulen O, Cai Q, Rubinsztein DC, Chi JT, Zhang DD, Li C, Toyokuni S, Liu J, Roh JL, Dai E, Juhasz G, Liu W, Zhang J, Yang M, Liu J, Zhu LQ, Zou W, Piacentini M, Ding WX, Yue Z, Xie Y, Petersen M, Gewirtz DA, Mandell MA, Chu CT, Sinha D, Eftekharpour E, Zhivotovsky B, Besteiro S, Gabrilovich DI, Kim DH, Kagan VE, Bayir H, Chen GC, Ayton S, Lünemann JD, Komatsu M, Krautwald S, Loos B, Baehrecke EH, Wang J, Lane JD, Sadoshima J, Yang WS, Gao M, Münz C, Thumm M, Kampmann M, Yu D, Lipinski MM, Jones JW, Jiang X, Zeh HJ, Kang R, Klionsky DJ, Kroemer G, Tang D. International consensus guidelines for the definition, detection, and interpretation of autophagy-dependent ferroptosis. Autophagy 2024; 20:1213-1246. [PMID: 38442890 PMCID: PMC11210914 DOI: 10.1080/15548627.2024.2319901] [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: 08/25/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 03/07/2024] Open
Abstract
Macroautophagy/autophagy is a complex degradation process with a dual role in cell death that is influenced by the cell types that are involved and the stressors they are exposed to. Ferroptosis is an iron-dependent oxidative form of cell death characterized by unrestricted lipid peroxidation in the context of heterogeneous and plastic mechanisms. Recent studies have shed light on the involvement of specific types of autophagy (e.g. ferritinophagy, lipophagy, and clockophagy) in initiating or executing ferroptotic cell death through the selective degradation of anti-injury proteins or organelles. Conversely, other forms of selective autophagy (e.g. reticulophagy and lysophagy) enhance the cellular defense against ferroptotic damage. Dysregulated autophagy-dependent ferroptosis has implications for a diverse range of pathological conditions. This review aims to present an updated definition of autophagy-dependent ferroptosis, discuss influential substrates and receptors, outline experimental methods, and propose guidelines for interpreting the results.Abbreviation: 3-MA:3-methyladenine; 4HNE: 4-hydroxynonenal; ACD: accidentalcell death; ADF: autophagy-dependentferroptosis; ARE: antioxidant response element; BH2:dihydrobiopterin; BH4: tetrahydrobiopterin; BMDMs: bonemarrow-derived macrophages; CMA: chaperone-mediated autophagy; CQ:chloroquine; DAMPs: danger/damage-associated molecular patterns; EMT,epithelial-mesenchymal transition; EPR: electronparamagnetic resonance; ER, endoplasmic reticulum; FRET: Försterresonance energy transfer; GFP: green fluorescent protein;GSH: glutathione;IF: immunofluorescence; IHC: immunohistochemistry; IOP, intraocularpressure; IRI: ischemia-reperfusion injury; LAA: linoleamide alkyne;MDA: malondialdehyde; PGSK: Phen Green™ SK;RCD: regulatedcell death; PUFAs: polyunsaturated fatty acids; RFP: red fluorescentprotein;ROS: reactive oxygen species; TBA: thiobarbituricacid; TBARS: thiobarbituric acid reactive substances; TEM:transmission electron microscopy.
Collapse
Affiliation(s)
- Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Andrey S. Tsvetkov
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Han-Ming Shen
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Ciro Isidoro
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | | | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Werner J.H. Koopman
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Shouqing Luo
- Peninsula Medical School, University of Plymouth, Plymouth, UK
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA Cancer-University of Liège, Liège, Belgium
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - David C. Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Donna D. Zhang
- Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shinya Toyokuni
- Department of Pathology and Biological Response, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Jinbao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Enyong Dai
- The Second Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Gabor Juhasz
- Biological Research Center, Institute of Genetics, Szeged, Hungary
- Department of Anatomy, Cell and Developmental Biology, Eotvos Lorand University, Budapest, Hungary
| | - Wei Liu
- Department of Orthopedics, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Minghua Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Pediatric Cancer, Changsha, China
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiping Zou
- Departments of Surgery and Pathology, University of Michigan Medical School, Ann Arbor, USA
| | - Mauro Piacentini
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
- National Institute for Infectious Diseases IRCCS “Lazzaro Spallanzani”, Rome, Italy
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yangchun Xie
- Department of Oncology, Central South University, Changsha, Hunan, China
| | - Morten Petersen
- Functional genomics, Department of Biology, Copenhagen University, Denmark
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA, USA
| | - Michael A. Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, USA
| | - Charleen T. Chu
- Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Wilmer Eye lnstitute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eftekhar Eftekharpour
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer, Villejuif, France; Gustave Roussy Cancer, Villejuif, France
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden, Europe
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Moscow, Russia
| | - Sébastien Besteiro
- LPHI, University Montpellier, CNRS, Montpellier, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | | | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Valerian E. Kagan
- Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York, USA
| | - Guang-Chao Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Scott Ayton
- Florey Institute, University of Melbourne, Parkville, Australia
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University School of Medicine, Bunkyo-ku Tokyo, Japan
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Eric H. Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Medical Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jon D. Lane
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Junichi Sadoshima
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine, Newark, USA
| | - Wan Seok Yang
- Department of Biological Sciences, St. John’s University, New York City, NY, USA
| | - Minghui Gao
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Christian Münz
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Michael Thumm
- Department of Cellular Biochemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Martin Kampmann
- Department of Biochemistry & Biophysics, University of California, San Francisco, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, USA
| | - Di Yu
- Faculty of Medicine, Frazer Institute, University of Queensland, Brisbane, Australia
- Faculty of Medicine, Ian Frazer Centre for Children’s Immunotherapy Research, Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - Marta M. Lipinski
- Department of Anesthesiology & Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jace W. Jones
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Herbert J. Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer, Villejuif, France; Gustave Roussy Cancer, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
50
|
Zhou X, Zhang D, Lei J, Ren J, Yang B, Cao Z, Guo C, Li Y. Polyphyllin I induces rapid ferroptosis in acute myeloid leukemia through simultaneous targeting PI3K/SREBP-1/SCD1 axis and triggering of lipid peroxidation. J Nat Med 2024; 78:618-632. [PMID: 38668832 DOI: 10.1007/s11418-024-01811-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/01/2024] [Indexed: 05/18/2024]
Abstract
Acute myeloid leukemia (AML) is a malignant disease that is difficult to completely cure. Polyphyllin I (PPI), a steroidal saponin isolated from Paris polyphylla, has exhibited multiple biological activities. Here, we discovered the superior cytotoxicity of PPI on AML cells MOLM-13 with an IC50 values of 0.44 ± 0.09 μM. Mechanically, PPI could cause ferroptosis via the accumulation of intracellular iron concentration and triggering lipid peroxidation. Interestingly, PPI could induced stronger ferroptosis in a short time of about 6 h compared to erastin. Furthermore, we demonstrate that PPI-induced rapid ferroptosis is due to the simultaneous targeting PI3K/SREBP-1/SCD1 axis and triggering lipid peroxidation, and PI3K inhibitor Alpelisib can enhance the activity of erastin-induced ferroptosis. Molecular docking simulations and kinase inhibition assays demonstrated that PPI is a PI3K inhibitor. In addition, PPI significantly inhibited tumor progression and prolonged mouse survival at 4 mg/kg with well tolerance. In summary, our study highlights the therapeutic potential of PPI for AML and shows its unique dual mechanism.
Collapse
Affiliation(s)
- Xinyu Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Duanna Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jieting Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jixia Ren
- College of Life Science, Liaocheng University, Liaocheng, 252059, China
| | - Bo Yang
- Department of Pharmacy, Panzhihua Central Hospital, Dali University, Panzhihua, China
- Department of Pharmacy, The Seventh People's Hospital of Chengdu, Chengdu, China
| | - Zhixing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Chuanjie Guo
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- Department of Pharmacy, The Seventh People's Hospital of Chengdu, Chengdu, China.
| | - Yuzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|