1
|
Liu Y, Stockwell BR, Jiang X, Gu W. p53-regulated non-apoptotic cell death pathways and their relevance in cancer and other diseases. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00842-3. [PMID: 40204927 DOI: 10.1038/s41580-025-00842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Programmed cell death is a mechanism that is crucial for numerous physiological and pathological processes. Whereas p53-mediated apoptosis is a major cell death pathway in cancer, accumulating evidence indicates that p53 also has crucial roles in controlling different non-apoptotic cell death (NACD) pathways, including ferroptosis, necroptosis, pyroptosis, autophagy-dependent cell death, entotic cell death, parthanatos and paraptosis, and may regulate PANoptosis, cuproptosis and disulfidptosis. Notably, the function of p53 in these NACDs substantially contributes to its biological effects, particularly in cancer development and other pathological processes. In this Review, we discuss recent advances in understanding the roles and underlying mechanisms of p53-mediated NACDs, focusing on ferroptosis, necroptosis and pyroptosis. We discuss the complex and distinct physiological settings in which NACDs are regulated by p53, and potential targeting of p53-regulated NACDs for the treatment of cancer and other human diseases. Finally, we highlight several important questions concerning p53-regulated NACDs that warrant further investigation.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, USA
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
2
|
Wu Z, Huang X, Lu X, Cao Y. The interplay of sleep deprivation, ferroptosis, and BACH1 in cardiovascular disease pathogenesis. Tissue Cell 2025; 95:102848. [PMID: 40081254 DOI: 10.1016/j.tice.2025.102848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/17/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND AND OBJECTIVE Sleep deprivation (SD) can induce cardiac dysfunction, myocardial injury, and sudden death. The purpose of this study is to investigate whether the mechanism of sleep deprivation induced myocardial injury is related to abnormal expression of BACH1 and ferroptosis of myocardial cells. METHODS Thirty-six male C57BL/6 J mice were used in the study. Twenty-four were exposed to SD through a modified multiple platform water method, while twelve served as controls. Cardiac function was assessed by small animal ultrasound super-resolution microimager and morphology by H&E staining. Ferroptosis model was established in H9c2 cardiomyocytes by Erastin. Biochemical assays measured key markers in serum and cell supernatants. TEM and flow cytometry evaluated mitochondrial changes and ROS levels. Western blot analyzed the expression of BACH1 and ferroptosis-related proteins. RESULTS Sleep deprivation caused increased heart rate (644 ± 30 bpm) and impaired cardiac function (EF 88.29 ± 3.97 %, FS 60.27 ± 2.70 %, LVIDd 3.14 ± 0.16 mm, LVIDs 1.30 ± 0.18 mm) in mice. It induced cardiomyocyte and mitochondrial damage, resulting in oxidative stress (MDA 7.16 ± 0.39 nmol/mg prot, GSH 33.88 ± 1.41 μg/mL, SOD 56.12 ± 1.44 U/mL, LDH 702.24 ± 33.48 U/L) and elevated CK levels (43.78 ± 2.30 U/mL). The expression of ANP, BACH1, and ferroptosis-related proteins (TFRC, SLC7A11, GPX4, NQO1, and HO-1) was altered. We observed aberrant oxidative stress indicators and expression of BACH1 and ferroptosis-related proteins in an Erastin-induced H9c2 cardiomyocytes model. CONCLUSIONS Sleep deprivation promotes BACH1 through ferroptosis and leads to myocardial injury, thereby revealing potential therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Zhenghua Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xuanxuan Huang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xinglong Lu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yin Cao
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
3
|
Wei X, He Y, Yu Y, Tang S, Liu R, Guo J, Jiang Q, Zhi X, Wang X, Meng D. The Multifaceted Roles of BACH1 in Disease: Implications for Biological Functions and Therapeutic Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412850. [PMID: 39887888 PMCID: PMC11905017 DOI: 10.1002/advs.202412850] [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: 10/13/2024] [Revised: 12/22/2024] [Indexed: 02/01/2025]
Abstract
BTB domain and CNC homolog 1 (BACH1) belongs to the family of basic leucine zipper proteins and is expressed in most mammalian tissues. It can regulate its own expression and play a role in transcriptionally activating or inhibiting downstream target genes. It has a crucial role in various biological processes, such as oxidative stress, cell cycle, heme homeostasis, and immune regulation. Recent research highlights BACH1's significant regulatory roles in a series of conditions, including stem cell pluripotency maintenance and differentiation, growth, senescence, and apoptosis. BACH1 is closely associated with cardiovascular diseases and contributes to angiogenesis, atherosclerosis, restenosis, pathological cardiac hypertrophy, myocardial infarction, and ischemia/reperfusion (I/R) injury. BACH1 promotes tumor cell proliferation and metastasis by altering tumor metabolism and the epithelial-mesenchymal transition phenotype. Moreover, BACH1 appears to show an adverse role in diseases such as neurodegenerative diseases, gastrointestinal disorders, leukemia, pulmonary fibrosis, and skin diseases. Inhibiting BACH1 may be beneficial for treating these diseases. This review summarizes the role of BACH1 and its regulatory mechanism in different cell types and diseases, proposing that precise targeted intervention of BACH1 may provide new strategies for human disease prevention and treatment.
Collapse
Affiliation(s)
- Xiangxiang Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Yunquan He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Yueyang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Sichong Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Ruiwen Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Jieyu Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Qingjun Jiang
- Department of Vascular & Endovascular Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Xinhong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| |
Collapse
|
4
|
Butera A, Amelio I. Deciphering the significance of p53 mutant proteins. Trends Cell Biol 2025; 35:258-268. [PMID: 38960851 DOI: 10.1016/j.tcb.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024]
Abstract
Mutations in the p53 gene compromise its role as guardian of genomic integrity, yielding predominantly missense p53 mutant proteins. The gain-of-function hypothesis has long suggested that these mutant proteins acquire new oncogenic properties; however, recent studies challenge this notion, indicating that targeting these mutants may not impact the fitness of cancer cells. Mounting evidence indicates that tumorigenesis involves a cooperative interplay between driver mutations and cellular state, influenced by developmental stage, external insults, and tissue damage. Consistently, the behavior and properties of p53 mutants are altered by the context. This article aims to provide a balanced summary of the evolving evidence regarding the contribution of p53 mutants in the biology of cancer while contemplating alternative frameworks to decipher the complexity of p53 mutants within their physiological contexts.
Collapse
Affiliation(s)
- Alessio Butera
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany
| | - Ivano Amelio
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
5
|
Liu J, Shen Y, Liu J, Xu D, Chang CY, Wang J, Zhou J, Haffty BG, Zhang L, Bargonetti J, De S, Hu W, Feng Z. Lipogenic enzyme FASN promotes mutant p53 accumulation and gain-of-function through palmitoylation. Nat Commun 2025; 16:1762. [PMID: 39971971 PMCID: PMC11839913 DOI: 10.1038/s41467-025-57099-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
The tumor-suppressive function of p53 is frequently disrupted by mutations in cancers. Missense mutant p53 (mutp53) protein often stabilizes and accumulates to high levels in cancers to promote tumorigenesis through the gain-of-function (GOF) mechanism. Currently, the mechanism of mutp53 accumulation and GOF is incompletely understood. Here, we identify the lipogenic enzyme FASN as an important regulator of mutp53 accumulation and GOF. FASN interacts with mutp53 to enhance mutp53 palmitoylation, which inhibits mutp53 ubiquitination to promote mutp53 accumulation and GOF. Blocking FASN genetically or by small-molecule inhibitors suppresses mutp53 palmitoylation to inhibit mutp53 accumulation, which in turn inhibits the growth of mutp53 tumors in orthotopic and subcutaneous xenograft tumor models and transgenic mice, as well as the growth of human tumor organoids carrying mutp53. Our results reveal that mutp53 palmitoylation is an important mechanism underlying mutp53 accumulation and GOF, and targeting FASN is a potential therapeutic strategy for cancers carrying mutp53.
Collapse
Affiliation(s)
- Juan Liu
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA
| | - Yiyun Shen
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA
| | - Jie Liu
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA
| | - Dandan Xu
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA
| | - Chun-Yuan Chang
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA
| | - Jianming Wang
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA
| | - Jason Zhou
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA
| | - Bruce G Haffty
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA
| | - Lanjing Zhang
- Department of Pathology, Princeton Medical Center, Princeton, NJ, USA
- Department of Cell Biology and Neuroscience, Rutgers-State University of New Jersey, Piscataway, NJ, USA
| | - Jill Bargonetti
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Subhajyoti De
- Center for Systems and Computational Biology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA.
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers-State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
6
|
Esmaeili A, Awasthi P, Tabaee S. Beyond immortality: Epstein-Barr virus and the intricate dance of programmed cell death in cancer development. Cancer Treat Res Commun 2025; 43:100880. [PMID: 39923321 DOI: 10.1016/j.ctarc.2025.100880] [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: 11/15/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
This comprehensive review delves into the intricate role of programmed cell death in Epstein-Barr virus (EBV)-associated malignancies, focusing on the sophisticated interplay between viral mechanisms and the host's immune response. The central objective is to unravel how EBV exerts control over cell death pathways such as apoptosis, ferroptosis, and autophagy, thereby fostering its persistence and oncogenic potential. By dissecting these mechanisms, the review seeks to identify therapeutic strategies that could disrupt EBV's manipulation of these pathways, enhancing immune recognition and opening new avenues for targeted treatment. A deeper understanding of the molecular underpinnings of EBV's influence on cell death not only enriches the field of viral oncology but also pinpoints targets for drug development. Furthermore, the insights gleaned from this review could catalyze the design of vaccines aimed at preventing EBV infection or curtailing its oncogenic impact. Innovatively, the review synthesizes recent discoveries on the multifaceted roles of non-coding RNAs and cellular signaling pathways in modulating cell death within the context of EBV infection. By consolidating current knowledge and identifying areas where understanding is lacking, it lays the groundwork for future research that could lead to significant advancements in vaccine development and therapeutic interventions for EBV-related cancers. This review underscores the critical necessity for ongoing investigation into the complex interplay between EBV and host cell death mechanisms, with the ultimate goal of enhancing patient outcomes in EBV-associated diseases.
Collapse
Affiliation(s)
- Arezoo Esmaeili
- Department of biology, Damghan Branch, Islamic Azad University, Damghan, Iran.
| | - Prankur Awasthi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Samira Tabaee
- Department of immunology, school of medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Xu Y, Xu X, Chai R, Wu X. Targeting ferroptosis to enhance the efficacy of mesenchymal stem cell-based treatments for intervertebral disc degeneration. Int J Biol Sci 2025; 21:1222-1241. [PMID: 39897051 PMCID: PMC11781166 DOI: 10.7150/ijbs.107021] [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: 11/15/2024] [Accepted: 01/11/2025] [Indexed: 02/04/2025] Open
Abstract
Although mesenchymal stromal cell (MSC) implantation shows promise for repairing intervertebral disc (IVD) degeneration (IVDD), their limited retention within degenerative IVDs compromises therapeutic efficacy. The oxidative stress in the microenvironment of degenerated IVDs induces a surge in reactive oxygen species production within MSCs, disrupting the balance between oxidation and antioxidation, and ultimately inducing ferroptosis. Recent evidence has suggested that targeting ferroptosis in MSCs could enhance MSC retention, extend the survival of transplanted MSCs, and markedly delay the pathological progression of IVDD. By targeting ferroptosis, a novel approach emerges to boost the efficacy of MSC transplantation therapy for IVDD. In this review, current research on targeting ferroptosis in MSCs is discussed from various perspectives, including the targeting of specific genes and pathways, drug preconditioning, and hydrogel encapsulation. A detailed discussion on the effects of targeting ferroptosis in MSCs on the transplantation repair of degenerated IVDs is provided. Insights that could guide improvements in stem cell transplantation therapies are also offered. Significantly, this review presents specific ideas for our future foundational research. These insights outline promising avenues for future clinical translation and will contribute to developing and optimizing treatment strategies for MSC transplantation therapy, maximizing benefits for patients with lumbar IVDD.
Collapse
Affiliation(s)
- Yuzhu Xu
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xuanfei Xu
- Department of Nuclear Medicine, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Renjie Chai
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xiaotao Wu
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, 210009, China
| |
Collapse
|
8
|
Su Z, Liu Y, Wang L, Gu W. Regulation of SLC7A11 as an unconventional checkpoint in tumorigenesis through ferroptosis. Genes Dis 2025; 12:101254. [PMID: 39569390 PMCID: PMC11577153 DOI: 10.1016/j.gendis.2024.101254] [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: 12/21/2023] [Revised: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 11/22/2024] Open
Abstract
Although cell-cycle arrest, senescence, and apoptosis are well accepted as the classic barriers in tumorigenesis, recent studies indicate that metabolic regulation is equally important as a major checkpoint in cancer development. It is well accepted that ferroptosis, an iron-dependent programmed cell death, acts as a new type of tumor suppression mechanism tightly linked with numerous metabolic pathways. SLC7A11 is a transmembrane cystine/glutamate transporter protein that plays a vital role in controlling ferroptosis in vivo. The levels of SLC7A11 are dynamically regulated by various types of stresses, such as oxidative stress, nutrient deprivation, endoplasmic reticulum stress, radiation, oncogenic stress, DNA damage, and immune stress. SLC7A11 can be transcriptionally regulated by both activators such as ATF4, NRF2, and ETS1, and repressors including BACH1, p53, ATF3, and STAT1 during stress responses. Moreover, SLC7A11 activity and its protein stability and cellular localization are also modulated upon stress. Patients' data show that SLC7A11 is overexpressed in various types of human cancers, and higher levels of SLC7A11 predict poorer overall survival. Growing evidence also suggests that targeting SLC7A11 is a promising approach in cancer therapy by effectively inhibiting tumor proliferation, invasion, and metastasis, as well as counteracting cancer stem cells and overcoming chemoresistance. This review highlights the regulation of SLC7A11 as an unconventional checkpoint in tumorigenesis through modulating ferroptotic responses under various types of stress.
Collapse
Affiliation(s)
- Zhenyi Su
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yanqing Liu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lin Wang
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
9
|
Gao Y, Siyu zhang, Zhang X, Du Y, Ni T, Hao S. Crosstalk between metabolic and epigenetic modifications during cell carcinogenesis. iScience 2024; 27:111359. [PMID: 39660050 PMCID: PMC11629229 DOI: 10.1016/j.isci.2024.111359] [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] [Indexed: 12/12/2024] Open
Abstract
Genetic mutations arising from various internal and external factors drive cells to become cancerous. Cancerous cells undergo numerous changes, including metabolic reprogramming and epigenetic modifications, to support their abnormal proliferation. This metabolic reprogramming leads to the altered expression of many metabolic enzymes and the accumulation of metabolites. Recent studies have shown that these enzymes and metabolites can serve as substrates or cofactors for chromatin-modifying enzymes, thereby participating in epigenetic modifications and promoting carcinogenesis. Additionally, epigenetic modifications play a role in the metabolic reprogramming and immune evasion of cancer cells, influencing cancer progression. This review focuses on the origins of cancer, particularly the metabolic reprogramming of cancer cells and changes in epigenetic modifications. We discuss how metabolites in cancer cells contribute to epigenetic remodeling, including lactylation, acetylation, succinylation, and crotonylation. Finally, we review the impact of epigenetic modifications on tumor immunity and the latest advancements in cancer therapies targeting these modifications.
Collapse
Affiliation(s)
- Yue Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Siyu zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Xianhong Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Yitian Du
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Ting Ni
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Shuailin Hao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| |
Collapse
|
10
|
Qiu L, Ma Z, Wu X. Mutant p53-Mediated Tumor Secretome: Bridging Tumor Cells and Stromal Cells. Genes (Basel) 2024; 15:1615. [PMID: 39766882 PMCID: PMC11675497 DOI: 10.3390/genes15121615] [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: 11/01/2024] [Revised: 12/06/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
The tumor secretome comprises the totality of protein factors secreted by various cell components within the tumor microenvironment, serving as the primary medium for signal transduction between tumor cells and between tumor cells and stromal cells. The deletion or mutation of the p53 gene leads to alterations in cellular secretion characteristics, contributing to the construction of the tumor microenvironment in a cell non-autonomous manner. This review discusses the critical roles of mutant p53 in regulating the tumor secretome to remodel the tumor microenvironment, drive tumor progression, and influence the plasticity of cancer-associated fibroblasts (CAFs) as well as the dynamics of tumor immunity by focusing on both secreted protein expression and secretion pathways. The aim is to provide new insights for targeted cancer therapies.
Collapse
Affiliation(s)
| | | | - Xiaoming Wu
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming 650500, China; (L.Q.); (Z.M.)
| |
Collapse
|
11
|
Liu J, Luo Y, Chen S, Wang G, Jin W, Jiang W, Li M, Wang Y, Yu J, Wei H, Zhang R, Zhou F, Ju L, Zhang Y, Xiao Y, Qian K, Wang X. Deubiquitylase USP52 Promotes Bladder Cancer Progression by Modulating Ferroptosis through Stabilizing SLC7A11/xCT. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403995. [PMID: 39392373 PMCID: PMC11615784 DOI: 10.1002/advs.202403995] [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: 04/16/2024] [Revised: 09/23/2024] [Indexed: 10/12/2024]
Abstract
Bladder cancer (BLCA) is a prevalent cancer with high case-fatality rates and a substantial economic burden worldwide. Understanding its molecular underpinnings to guide clinical management is crucial. Ferroptosis, a recently described non-apoptotic form of cell death, is initiated by the lethal accumulation of iron-dependent lipid peroxidation products. Despite growing interest, the roles and vulnerabilities determining ferroptosis sensitivity in BLCA remain unclear. Re-analysis of single-cell RNA data reveals a decrease in high-ferroptosis cancer cells as BLCA advances. USP52/PAN2 is identified as a key regulator of ferroptosis in BLCA through an unbiased siRNA screen targeting 96 deubiquitylases (DUBs). Functionally, USP52 depletion impedes glutathione (GSH) synthesis by promoting xCT protein degradation, increasing lipid peroxidation and ferroptosis susceptibility, thus suppressing BLCA progression. Mechanistically, USP52 interacts with xCT and enzymatically cleaves the K48-conjugated ubiquitin chains at K4 and K12, enhancing its protein stability. Clinical BLCA samples demonstrate a positive correlation between USP52 and xCT expression, with high USP52 levels associated with aggressive disease progression and poor prognosis. In vivo, USP52 depletion combined with ferroptosis triggers imidazole ketone Erastin (IKE) synergistically restrains BLCA progression by inducing ferroptosis. These findings elucidate the role of the USP52-xCT axis in BLCA and highlight the therapeutic potential of targeting USP52 and ferroptosis inducers in BLCA.
Collapse
Affiliation(s)
- Jianmin Liu
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Yongwen Luo
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Siming Chen
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Gang Wang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Biological RepositoriesHuman Genetic Resources Preservation Center of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Wan Jin
- Department of Biological RepositoriesHuman Genetic Resources Preservation Center of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Urological DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Euler TechnologyZGC Life Sciences ParkBeijing102206China
| | - Wenyu Jiang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Mingxing Li
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Yejinpeng Wang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Jingtian Yu
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Houyi Wei
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Renjie Zhang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Fenfang Zhou
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of RadiologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Lingao Ju
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Biological RepositoriesHuman Genetic Resources Preservation Center of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Yi Zhang
- Department of Biological RepositoriesHuman Genetic Resources Preservation Center of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
- Euler TechnologyZGC Life Sciences ParkBeijing102206China
| | - Yu Xiao
- Department of Biological RepositoriesHuman Genetic Resources Preservation Center of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Urological DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Kaiyu Qian
- Department of Biological RepositoriesHuman Genetic Resources Preservation Center of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Xinghuan Wang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhan430071China
- Wuhan Research Center for Infectious Diseases and CancerChinese Academy of Medical SciencesWuhan430071China
- Medical Research InstituteFrontier Science Center for Immunology and MetabolismTaikang Center for Life and Medical SciencesWuhan UniversityWuhan430071China
| |
Collapse
|
12
|
Ge A, Xiang W, Li Y, Zhao D, Chen J, Daga P, Dai CC, Yang K, Yan Y, Hao M, Zhang B, Xiao W. Broadening horizons: the multifaceted role of ferroptosis in breast cancer. Front Immunol 2024; 15:1455741. [PMID: 39664391 PMCID: PMC11631881 DOI: 10.3389/fimmu.2024.1455741] [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: 06/27/2024] [Accepted: 10/21/2024] [Indexed: 12/13/2024] Open
Abstract
Breast cancer poses a serious threat to women's health globally. Current radiotherapy and chemotherapy regimens can induce drug-resistance effects in cancer tissues, such as anti-apoptosis, anti-pyroptosis, and anti-necroptosis, leading to poor clinical outcomes in the treatment of breast cancer. Ferroptosis is a novel programmed cell death modality characterized by iron overload, excessive generation of reactive oxygen species, and membrane lipid peroxidation. The occurrence of ferroptosis results from the imbalance between intracellular peroxidation mechanisms (executive system) and antioxidant mechanisms (defensive system), specifically involving iron metabolism pathways, amino acid metabolism pathways, and lipid metabolism pathways. In recent years, it has been found that ferroptosis is associated with the progression of various diseases, including tumors, hypertension, diabetes, and Alzheimer's disease. Studies have confirmed that triggering ferroptosis in breast cancer cells can significantly inhibit cancer cell proliferation and invasion, and improve cancer cell sensitivity to radiotherapy and chemotherapy, making induction of ferroptosis a potential strategy for the treatment of breast cancer. This paper reviews the development of the concept of ferroptosis, the mechanisms of ferroptosis (including signaling pathways such as GSH-GPX4, FSP1-CoQ1, DHODH-CoQ10, and GCH1-BH4) in breast cancer disease, the latest research progress, and summarizes the research on ferroptosis in breast cancer disease within the framework of metabolism, reactive oxygen biology, and iron biology. The key regulatory factors and mechanisms of ferroptosis in breast cancer disease, as well as important concepts and significant open questions in the field of ferroptosis and related natural compounds, are introduced. It is hoped that future research will make further breakthroughs in the regulatory mechanisms of ferroptosis and the use of ferroptosis in treating breast cancer cells. Meanwhile, natural compounds may also become a new direction for potential drug development targeting ferroptosis in breast cancer treatment. This provides a theoretical basis and opens up a new pathway for research and the development of drugs for the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wang Xiang
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| | - Yan Li
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Da Zhao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junpeng Chen
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, United States
- Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China
| | - Pawan Daga
- Department of Internal Medicine, University of Louisville, Louisville, KY, United States
| | - Charles C. Dai
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States
| | - Kailin Yang
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yexing Yan
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | | | - Wei Xiao
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| |
Collapse
|
13
|
Nakamura T, Conrad M. Exploiting ferroptosis vulnerabilities in cancer. Nat Cell Biol 2024; 26:1407-1419. [PMID: 38858502 DOI: 10.1038/s41556-024-01425-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/17/2024] [Indexed: 06/12/2024]
Abstract
Ferroptosis is a distinct lipid peroxidation-dependent form of necrotic cell death. This process has been increasingly contemplated as a new target for cancer therapy because of an intrinsic or acquired ferroptosis vulnerability in difficult-to-treat cancers and tumour microenvironments. Here we review recent advances in our understanding of the molecular mechanisms that underlie ferroptosis and highlight available tools for the modulation of ferroptosis sensitivity in cancer cells and communication with immune cells within the tumour microenvironment. We further discuss how these new insights into ferroptosis-activating pathways can become new armouries in the fight against cancer.
Collapse
Affiliation(s)
- Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets & Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets & Therapeutics Center, Helmholtz Munich, Neuherberg, Germany.
| |
Collapse
|
14
|
Mao C, Wang M, Zhuang L, Gan B. Metabolic cell death in cancer: ferroptosis, cuproptosis, disulfidptosis, and beyond. Protein Cell 2024; 15:642-660. [PMID: 38428031 PMCID: PMC11365558 DOI: 10.1093/procel/pwae003] [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: 01/05/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024] Open
Abstract
Cell death resistance represents a hallmark of cancer. Recent studies have identified metabolic cell death as unique forms of regulated cell death resulting from an imbalance in the cellular metabolism. This review discusses the mechanisms of metabolic cell death-ferroptosis, cuproptosis, disulfidptosis, lysozincrosis, and alkaliptosis-and explores their potential in cancer therapy. Our review underscores the complexity of the metabolic cell death pathways and offers insights into innovative therapeutic avenues for cancer treatment.
Collapse
Affiliation(s)
- Chao Mao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Min Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
15
|
Dai E, Chen X, Linkermann A, Jiang X, Kang R, Kagan VE, Bayir H, Yang WS, Garcia-Saez AJ, Ioannou MS, Janowitz T, Ran Q, Gu W, Gan B, Krysko DV, Zhu X, Wang J, Krautwald S, Toyokuni S, Xie Y, Greten FR, Yi Q, Schick J, Liu J, Gabrilovich DI, Liu J, Zeh HJ, Zhang DD, Yang M, Iovanna J, Kopf M, Adolph TE, Chi JT, Li C, Ichijo H, Karin M, Sankaran VG, Zou W, Galluzzi L, Bush AI, Li B, Melino G, Baehrecke EH, Lotze MT, Klionsky DJ, Stockwell BR, Kroemer G, Tang D. A guideline on the molecular ecosystem regulating ferroptosis. Nat Cell Biol 2024; 26:1447-1457. [PMID: 38424270 PMCID: PMC11650678 DOI: 10.1038/s41556-024-01360-8] [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: 09/01/2023] [Accepted: 01/18/2024] [Indexed: 03/02/2024]
Abstract
Ferroptosis, an intricately regulated form of cell death characterized by uncontrolled lipid peroxidation, has garnered substantial interest since this term was first coined in 2012. Recent years have witnessed remarkable progress in elucidating the detailed molecular mechanisms that govern ferroptosis induction and defence, with particular emphasis on the roles of heterogeneity and plasticity. In this Review, we discuss the molecular ecosystem of ferroptosis, with implications that may inform and enable safe and effective therapeutic strategies across a broad spectrum of diseases.
Collapse
Affiliation(s)
- Enyong Dai
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rui Kang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Valerian E Kagan
- Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Wan Seok Yang
- Department of Biological Sciences, St. John's University, New York, NY, USA
| | - Ana J Garcia-Saez
- Institute for Genetics, CECAD, University of Cologne, Cologne, Germany
| | - Maria S Ioannou
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | | | - Qitao Ran
- Department of Cell Systems and Anatomy, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Xiaofeng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, and Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital and College of Medical Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| | - 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
| | - Yangchun Xie
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Florian R Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Qing Yi
- Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
| | - Joel Schick
- Genetics and Cellular Engineering Group, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Jinbao Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Herbert J Zeh
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Minghua Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Research Center of Pediatric Cancer, Changsha, China
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology Center for Applied Genomic Technologies, Duke University, Durham, NC, USA
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Weiping Zou
- Departments of Surgery and Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - 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
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Binghui Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Department of Cancer Cell Biology and National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Gerry Melino
- Department of Experimental Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Michael T Lotze
- Departments of Surgery, Immunology and Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Brent R Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA.
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, 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 Campus, Villejuif, France.
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Daolin Tang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
16
|
Berndt C, Alborzinia H, Amen VS, Ayton S, Barayeu U, Bartelt A, Bayir H, Bebber CM, Birsoy K, Böttcher JP, Brabletz S, Brabletz T, Brown AR, Brüne B, Bulli G, Bruneau A, Chen Q, DeNicola GM, Dick TP, Distéfano A, Dixon SJ, Engler JB, Esser-von Bieren J, Fedorova M, Friedmann Angeli JP, Friese MA, Fuhrmann DC, García-Sáez AJ, Garbowicz K, Götz M, Gu W, Hammerich L, Hassannia B, Jiang X, Jeridi A, Kang YP, Kagan VE, Konrad DB, Kotschi S, Lei P, Le Tertre M, Lev S, Liang D, Linkermann A, Lohr C, Lorenz S, Luedde T, Methner A, Michalke B, Milton AV, Min J, Mishima E, Müller S, Motohashi H, Muckenthaler MU, Murakami S, Olzmann JA, Pagnussat G, Pan Z, Papagiannakopoulos T, Pedrera Puentes L, Pratt DA, Proneth B, Ramsauer L, Rodriguez R, Saito Y, Schmidt F, Schmitt C, Schulze A, Schwab A, Schwantes A, Soula M, Spitzlberger B, Stockwell BR, Thewes L, Thorn-Seshold O, Toyokuni S, Tonnus W, Trumpp A, Vandenabeele P, Vanden Berghe T, Venkataramani V, Vogel FCE, von Karstedt S, Wang F, Westermann F, Wientjens C, Wilhelm C, Wölk M, Wu K, Yang X, Yu F, Zou Y, Conrad M. Ferroptosis in health and disease. Redox Biol 2024; 75:103211. [PMID: 38908072 PMCID: PMC11253697 DOI: 10.1016/j.redox.2024.103211] [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: 03/21/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
Ferroptosis is a pervasive non-apoptotic form of cell death highly relevant in various degenerative diseases and malignancies. The hallmark of ferroptosis is uncontrolled and overwhelming peroxidation of polyunsaturated fatty acids contained in membrane phospholipids, which eventually leads to rupture of the plasma membrane. Ferroptosis is unique in that it is essentially a spontaneous, uncatalyzed chemical process based on perturbed iron and redox homeostasis contributing to the cell death process, but that it is nonetheless modulated by many metabolic nodes that impinge on the cells' susceptibility to ferroptosis. Among the various nodes affecting ferroptosis sensitivity, several have emerged as promising candidates for pharmacological intervention, rendering ferroptosis-related proteins attractive targets for the treatment of numerous currently incurable diseases. Herein, the current members of a Germany-wide research consortium focusing on ferroptosis research, as well as key external experts in ferroptosis who have made seminal contributions to this rapidly growing and exciting field of research, have gathered to provide a comprehensive, state-of-the-art review on ferroptosis. Specific topics include: basic mechanisms, in vivo relevance, specialized methodologies, chemical and pharmacological tools, and the potential contribution of ferroptosis to disease etiopathology and progression. We hope that this article will not only provide established scientists and newcomers to the field with an overview of the multiple facets of ferroptosis, but also encourage additional efforts to characterize further molecular pathways modulating ferroptosis, with the ultimate goal to develop novel pharmacotherapies to tackle the various diseases associated with - or caused by - ferroptosis.
Collapse
Affiliation(s)
- Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Vera Skafar Amen
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - Uladzimir Barayeu
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany; Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany; German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York City, NY, USA
| | - Christina M Bebber
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Kivanc Birsoy
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Ashley R Brown
- Department of Biological Sciences, Columbia University, New York City, NY, USA
| | - Bernhard Brüne
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Giorgia Bulli
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany
| | - Alix Bruneau
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Gina M DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Ayelén Distéfano
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jan B Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | | | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | - Dominic C Fuhrmann
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Ana J García-Sáez
- Institute for Genetics, CECAD, University of Cologne, Germany; Max Planck Institute of Biophysics, Frankfurt/Main, Germany
| | | | - Magdalena Götz
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, Germany
| | - Wei Gu
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | | | - Xuejun Jiang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Aicha Jeridi
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), Germany, Member of the German Center for Lung Research (DZL)
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Republic of Korea
| | | | - David B Konrad
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Peng Lei
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Marlène Le Tertre
- Center for Translational Biomedical Iron Research, Heidelberg University, Germany
| | - Sima Lev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Deguang Liang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany; Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Carolin Lohr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Svenja Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Axel Methner
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Bernhard Michalke
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Germany
| | - Anna V Milton
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Junxia Min
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | | | - Shohei Murakami
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Gabriela Pagnussat
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Zijan Pan
- School of Life Sciences, Westlake University, Hangzhou, China
| | | | | | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Canada
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Lukas Ramsauer
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | | | - Yoshiro Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Felix Schmidt
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Carina Schmitt
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Almut Schulze
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Annemarie Schwab
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Anna Schwantes
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Mariluz Soula
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Benedikt Spitzlberger
- Department of Immunobiology, Université de Lausanne, Switzerland; Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York City, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Chemistry, Columbia University, New York, NY, USA
| | - Leonie Thewes
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Nagoya, Japan
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Department of Biomedical Sciences, University of Antwerp, Belgium; VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Vivek Venkataramani
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Germany
| | - Felix C E Vogel
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silvia von Karstedt
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Germany
| | - Fudi Wang
- School of Medicine, Zhejiang University, Hangzhou, China
| | | | - Chantal Wientjens
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - Katherine Wu
- Department of Pathology, Grossman School of Medicine, New York University, NY, USA
| | - Xin Yang
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Fan Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yilong Zou
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany.
| |
Collapse
|
17
|
Murray MB, Dixon SJ. Ferroptosis regulation by Cap'n'collar family transcription factors. J Biol Chem 2024; 300:107583. [PMID: 39025451 PMCID: PMC11387702 DOI: 10.1016/j.jbc.2024.107583] [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: 03/29/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024] Open
Abstract
Ferroptosis is an iron-dependent cell death mechanism that may be important to prevent tumor formation and useful as a target for new cancer therapies. Transcriptional networks play a crucial role in shaping ferroptosis sensitivity by regulating the expression of transporters, metabolic enzymes, and other proteins. The Cap'n'collar (CNC) protein NFE2 like bZIP transcription factor 2 (NFE2L2, also known as NRF2) is a key regulator of ferroptosis in many cells and contexts. Emerging evidence indicates that the related CNC family members, BTB domain and CNC homolog 1 (BACH1) and NFE2 like bZIP transcription factor 1 (NFE2L1), also have roles in ferroptosis regulation. Here, we comprehensively review the role of CNC transcription factors in governing cellular sensitivity to ferroptosis. We describe how CNC family members regulate ferroptosis sensitivity through modulation of iron, lipid, and redox metabolism. We also use examples of ferroptosis regulation by CNC proteins to illustrate the flexible and highly context-dependent nature of the ferroptosis mechanism in different cells and conditions.
Collapse
Affiliation(s)
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, California, USA.
| |
Collapse
|
18
|
Nishizawa H, Matsumoto M, Yamanaka M, Irikura R, Nakajima K, Tada K, Nakayama Y, Konishi M, Itoh N, Funayama R, Nakayama K, Igarashi K. BACH1 inhibits senescence, obesity, and short lifespan by ferroptotic FGF21 secretion. Cell Rep 2024; 43:114403. [PMID: 38943639 DOI: 10.1016/j.celrep.2024.114403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/14/2024] [Accepted: 06/11/2024] [Indexed: 07/01/2024] Open
Abstract
Ferroptosis is a type of regulated cell death characterized by iron-dependent lipid peroxidation. A model cell system is constructed to induce ferroptosis by re-expressing the transcription factor BACH1, a potent ferroptosis inducer, in immortalized mouse embryonic fibroblasts (iMEFs). The transfer of the culture supernatant from ferroptotic iMEFs activates the proliferation of hepatoma cells and other fibroblasts and suppresses cellular senescence-like features. The BACH1-dependent secretion of the longevity factor FGF21 is increased in ferroptotic iMEFs. The anti-senescent effects of the culture supernatant from these iMEFs are abrogated by Fgf21 knockout. BACH1 activates the transcription of Fgf21 by promoting ferroptotic stress and increases FGF21 protein expression by suppressing its autophagic degradation through transcriptional Sqstm1 and Lamp2 repression. The BACH1-induced ferroptotic FGF21 secretion suppresses obesity in high-fat diet-fed mice and the short lifespan of progeria mice. The inhibition of these aging-related phenotypes can be physiologically significant regarding ferroptosis.
Collapse
Affiliation(s)
- Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan.
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Gladstone Institute of Neurological Disease, Gladstone Institute, San Francisco, CA 94158, USA
| | - Riko Irikura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Kazuma Nakajima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Keisuke Tada
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Yoshiaki Nakayama
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Hyogo 658-8558, Japan
| | - Morichika Konishi
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Hyogo 658-8558, Japan
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo, Kyoto 606-8501, Japan
| | - Ryo Funayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Keiko Nakayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan.
| |
Collapse
|
19
|
He S, Luo C, Shi F, Zhou J, Shang L. The Emerging Role of Ferroptosis in EBV-Associated Cancer: Implications for Cancer Therapy. BIOLOGY 2024; 13:543. [PMID: 39056735 PMCID: PMC11274159 DOI: 10.3390/biology13070543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Ferroptosis is a novel and iron-dependent form of programmed cell death, which has been implicated in the pathogenesis of various human cancers. EBV is a well-recognized oncogenic virus that controls multiple signaling pathways within the host cell, including ferroptosis signaling. Recent studies show that inducing ferroptosis could be an efficient therapeutic strategy for EBV-associated tumors. This review will firstly describe the mechanism of ferroptosis, then summarize EBV infection and EBV-associated tumors, as well as the crosstalk between EBV infection and the ferroptosis signaling pathway, and finally discuss the role and potential application of ferroptosis-related reagents in EBV-associated tumors.
Collapse
Affiliation(s)
- Shan He
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha 410078, China; (S.H.); (C.L.); (F.S.); (J.Z.)
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/XiangYa Hospital, Central South University, Changsha 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Cheng Luo
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha 410078, China; (S.H.); (C.L.); (F.S.); (J.Z.)
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/XiangYa Hospital, Central South University, Changsha 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha 410078, China; (S.H.); (C.L.); (F.S.); (J.Z.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Jianhua Zhou
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha 410078, China; (S.H.); (C.L.); (F.S.); (J.Z.)
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/XiangYa Hospital, Central South University, Changsha 410078, China
| | - Li Shang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha 410078, China; (S.H.); (C.L.); (F.S.); (J.Z.)
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/XiangYa Hospital, Central South University, Changsha 410078, China
| |
Collapse
|
20
|
Liu Y, Su Z, Tavana O, Gu W. Understanding the complexity of p53 in a new era of tumor suppression. Cancer Cell 2024; 42:946-967. [PMID: 38729160 PMCID: PMC11190820 DOI: 10.1016/j.ccell.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/15/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024]
Abstract
p53 was discovered 45 years ago as an SV40 large T antigen binding protein, coded by the most frequently mutated TP53 gene in human cancers. As a transcription factor, p53 is tightly regulated by a rich network of post-translational modifications to execute its diverse functions in tumor suppression. Although early studies established p53-mediated cell-cycle arrest, apoptosis, and senescence as the classic barriers in cancer development, a growing number of new functions of p53 have been discovered and the scope of p53-mediated anti-tumor activity is largely expanded. Here, we review the complexity of different layers of p53 regulation, and the recent advance of the p53 pathway in metabolism, ferroptosis, immunity, and others that contribute to tumor suppression. We also discuss the challenge regarding how to activate p53 function specifically effective in inhibiting tumor growth without harming normal homeostasis for cancer therapy.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zhenyi Su
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Omid Tavana
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
21
|
Su Z, Liu Y, Xia Z, Rustgi AK, Gu W. An unexpected role for the ketogenic diet in triggering tumor metastasis by modulating BACH1-mediated transcription. SCIENCE ADVANCES 2024; 10:eadm9481. [PMID: 38838145 PMCID: PMC11152127 DOI: 10.1126/sciadv.adm9481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/30/2024] [Indexed: 06/07/2024]
Abstract
We have found that the ketogenic (Keto) diet is able to, unexpectedly, promote the metastatic potential of cancer cells in complementary mouse models. Notably, the Keto diet-induced tumor metastasis is dependent on BTB domain and CNC homolog 1 (BACH1) and its up-regulation of pro-metastatic targets, including cell migration-inducing hyaluronidase 1, in response to the Keto diet. By contrast, upon genetic knockout or pharmacological inhibition of endogenous BACH1, the Keto diet-mediated activation of those targets is largely diminished, and the effects on tumor metastasis are completely abolished. Mechanistically, upon administration of the Keto diet, the levels of activating transcription factor 4 (ATF4) are markedly induced. Through direct interaction with BACH1, ATF4 is recruited to those pro-metastatic target promoters and enhances BACH1-mediated transcriptional activation. Together, these data implicate a distinct transcription regulatory program of BACH1 for tumor metastasis induced by the Keto diet. Our study also raises a potential health risk of the Keto diet in human patients with cancer.
Collapse
Affiliation(s)
- Zhenyi Su
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| | - Yanqing Liu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| | - Zhangchuan Xia
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032 USA
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| |
Collapse
|
22
|
Chen F, Kang R, Tang D, Liu J. Ferroptosis: principles and significance in health and disease. J Hematol Oncol 2024; 17:41. [PMID: 38844964 PMCID: PMC11157757 DOI: 10.1186/s13045-024-01564-3] [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] [Accepted: 06/02/2024] [Indexed: 06/09/2024] Open
Abstract
Ferroptosis, an iron-dependent form of cell death characterized by uncontrolled lipid peroxidation, is governed by molecular networks involving diverse molecules and organelles. Since its recognition as a non-apoptotic cell death pathway in 2012, ferroptosis has emerged as a crucial mechanism in numerous physiological and pathological contexts, leading to significant therapeutic advancements across a wide range of diseases. This review summarizes the fundamental molecular mechanisms and regulatory pathways underlying ferroptosis, including both GPX4-dependent and -independent antioxidant mechanisms. Additionally, we examine the involvement of ferroptosis in various pathological conditions, including cancer, neurodegenerative diseases, sepsis, ischemia-reperfusion injury, autoimmune disorders, and metabolic disorders. Specifically, we explore the role of ferroptosis in response to chemotherapy, radiotherapy, immunotherapy, nanotherapy, and targeted therapy. Furthermore, we discuss pharmacological strategies for modulating ferroptosis and potential biomarkers for monitoring this process. Lastly, we elucidate the interplay between ferroptosis and other forms of regulated cell death. Such insights hold promise for advancing our understanding of ferroptosis in the context of human health and disease.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, 75390, USA.
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China.
| |
Collapse
|
23
|
He B, Bie Q, Zhao R, Yan Y, Dong G, Zhang B, Wang S, Xu W, Tian D, Hao Y, Zhang Y, Zhao M, Xiong H, Zhang B. Arachidonic acid released by PIK3CA mutant tumor cells triggers malignant transformation of colonic epithelium by inducing chromatin remodeling. Cell Rep Med 2024; 5:101510. [PMID: 38614093 PMCID: PMC11148513 DOI: 10.1016/j.xcrm.2024.101510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/07/2024] [Accepted: 03/20/2024] [Indexed: 04/15/2024]
Abstract
Key gene mutations are essential for colorectal cancer (CRC) development; however, how the mutated tumor cells impact the surrounding normal cells to promote tumor progression has not been well defined. Here, we report that PIK3CA mutant tumor cells transmit oncogenic signals and result in malignant transformation of intestinal epithelial cells (IECs) via paracrine exosomal arachidonic acid (AA)-induced H3K4 trimethylation. Mechanistically, PIK3CA mutations sustain SGK3-FBW7-mediated stability of the cPLA2 protein, leading to the synthetic increase in AA, which is transported through exosome and accumulated in IECs. Transferred AA directly binds Menin and strengthens the interactions of Menin and MLL1/2 methyltransferase. Finally, the combination of VTP50469, an inhibitor of the Menin-MLL interaction, and alpelisib synergistically represses PDX tumors harboring PIK3CA mutations. Together, these findings unveil the metabolic link between PIK3CA mutant tumor cells and the IECs, highlighting AA as the potential target for the treatment of patients with CRC harboring PIK3CA mutations.
Collapse
Affiliation(s)
- Baoyu He
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, China; School of Integrative Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Qingli Bie
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, China; School of Integrative Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Rou Zhao
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, China
| | - Yugang Yan
- School of Medical Engineering, Jining Medical University, Jining, Shandong 272067, China
| | - Guanjun Dong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, China
| | - Baogui Zhang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, China
| | - Sen Wang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, China
| | - Wenrong Xu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Dongxing Tian
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, China
| | - Yujun Hao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Yanhua Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Mingsheng Zhao
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, China.
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, China.
| |
Collapse
|
24
|
Diao J, Jia Y, Dai E, Liu J, Kang R, Tang D, Han L, Zhong Y, Meng L. Ferroptotic therapy in cancer: benefits, side effects, and risks. Mol Cancer 2024; 23:89. [PMID: 38702722 PMCID: PMC11067110 DOI: 10.1186/s12943-024-01999-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/10/2024] [Indexed: 05/06/2024] Open
Abstract
Ferroptosis is a type of regulated cell death characterized by iron accumulation and uncontrolled lipid peroxidation, leading to plasma membrane rupture and intracellular content release. Originally investigated as a targeted therapy for cancer cells carrying oncogenic RAS mutations, ferroptosis induction now exhibits potential to complement chemotherapy, immunotherapy, and radiotherapy in various cancer types. However, it can lead to side effects, including immune cell death, bone marrow impairment, liver and kidney damage, cachexia (severe weight loss and muscle wasting), and secondary tumorigenesis. In this review, we discuss the advantages and offer an overview of the diverse range of documented side effects. Furthermore, we examine the underlying mechanisms and explore potential strategies for side effect mitigation.
Collapse
Affiliation(s)
- Jiandong Diao
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Yuanyuan Jia
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Enyong Dai
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Jiao Liu
- DAMP laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Rui Kang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daolin Tang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Leng Han
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China.
| | - Yingjie Zhong
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China.
| | - Lingjun Meng
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China.
| |
Collapse
|
25
|
Pang Q, Tang Z, Luo L. The crosstalk between oncogenic signaling and ferroptosis in cancer. Crit Rev Oncol Hematol 2024; 197:104349. [PMID: 38626848 DOI: 10.1016/j.critrevonc.2024.104349] [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: 09/30/2023] [Revised: 03/13/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Ferroptosis, a novel form of cell death regulation, was identified in 2012. It is characterized by unique features that differentiate it from other types of cell death, including necrosis, apoptosis, autophagy, and pyroptosis. Ferroptosis is defined by an abundance of iron ions and lipid peroxidation, resulting in alterations in subcellular structures, an elevation in reactive oxygen species (ROS), a reduction in glutathione (GSH) levels, and an augmentation in Fe (II) cytokines. Ferroptosis, a regulated process, is controlled by an intricate network of signaling pathways, where multiple stimuli can either enhance or hinder the process. This review primarily examines the defensive mechanisms of ferroptosis and its interaction with the tumor microenvironment. The analysis focuses on the pathways that involve AMPK, p53, NF2, mTOR, System Xc-, Wnt, Hippo, Nrf2, and cGAS-STING. The text discusses the possibilities of employing a combination therapy that targets several pathways for the treatment of cancer. It emphasizes the necessity for additional study in this field.
Collapse
Affiliation(s)
- Qianghu Pang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Zhirou Tang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang,School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
26
|
Lei G, Zhuang L, Gan B. The roles of ferroptosis in cancer: Tumor suppression, tumor microenvironment, and therapeutic interventions. Cancer Cell 2024; 42:513-534. [PMID: 38593779 DOI: 10.1016/j.ccell.2024.03.011] [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: 11/30/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
In cancer treatment, the recurrent challenge of inducing apoptosis through conventional therapeutic modalities, often thwarted by therapy resistance, emphasizes the critical need to explore alternative cell death pathways. Ferroptosis, an iron-dependent form of regulated cell death triggered by the lethal accumulation of lipid peroxides on cellular membranes, has emerged as one such promising frontier in oncology. Induction of ferroptosis not only suppresses tumor growth but also holds potential for augmenting immunotherapy responses and surmounting resistance to existing cancer therapies. This review navigates the role of ferroptosis in tumor suppression. Furthermore, we delve into the complex role of ferroptosis within the tumor microenvironment and its interplay with antitumor immunity, offering insights into the prospect of targeting ferroptosis as a strategic approach in cancer therapy.
Collapse
Affiliation(s)
- Guang Lei
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
27
|
Yang X, Wang Z, Samovich SN, Kapralov AA, Amoscato AA, Tyurin VA, Dar HH, Li Z, Duan S, Kon N, Chen D, Tycko B, Zhang Z, Jiang X, Bayir H, Stockwell BR, Kagan VE, Gu W. PHLDA2-mediated phosphatidic acid peroxidation triggers a distinct ferroptotic response during tumor suppression. Cell Metab 2024; 36:762-777.e9. [PMID: 38309267 PMCID: PMC11209835 DOI: 10.1016/j.cmet.2024.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/14/2023] [Accepted: 01/10/2024] [Indexed: 02/05/2024]
Abstract
Although the role of ferroptosis in killing tumor cells is well established, recent studies indicate that ferroptosis inducers also sabotage anti-tumor immunity by killing neutrophils and thus unexpectedly stimulate tumor growth, raising a serious issue about whether ferroptosis effectively suppresses tumor development in vivo. Through genome-wide CRISPR-Cas9 screenings, we discover a pleckstrin homology-like domain family A member 2 (PHLDA2)-mediated ferroptosis pathway that is neither ACSL4-dependent nor requires common ferroptosis inducers. PHLDA2-mediated ferroptosis acts through the peroxidation of phosphatidic acid (PA) upon high levels of reactive oxygen species (ROS). ROS-induced ferroptosis is critical for tumor growth in the absence of common ferroptosis inducers; strikingly, loss of PHLDA2 abrogates ROS-induced ferroptosis and promotes tumor growth but has no obvious effect in normal tissues in both immunodeficient and immunocompetent mouse tumor models. These data demonstrate that PHLDA2-mediated PA peroxidation triggers a distinct ferroptosis response critical for tumor suppression and reveal that PHLDA2-mediated ferroptosis occurs naturally in vivo without any treatment from ferroptosis inducers.
Collapse
Affiliation(s)
- Xin Yang
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Zhe Wang
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Svetlana N Samovich
- Center for Free Radical and Antioxidant Health and Departments of Environmental Health, Chemistry, Pharmacology and Chemical Biology, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alexander A Kapralov
- Center for Free Radical and Antioxidant Health and Departments of Environmental Health, Chemistry, Pharmacology and Chemical Biology, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Andrew A Amoscato
- Center for Free Radical and Antioxidant Health and Departments of Environmental Health, Chemistry, Pharmacology and Chemical Biology, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Vladimir A Tyurin
- Center for Free Radical and Antioxidant Health and Departments of Environmental Health, Chemistry, Pharmacology and Chemical Biology, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Haider H Dar
- Center for Free Radical and Antioxidant Health and Departments of Environmental Health, Chemistry, Pharmacology and Chemical Biology, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Zhiming Li
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Shoufu Duan
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Ning Kon
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Delin Chen
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Benjamin Tycko
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ 07110, USA
| | - Zhiguo Zhang
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA; Department of Pediatrics and Department of Genetics and Development, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Hülya Bayir
- Center for Free Radical and Antioxidant Health and Departments of Environmental Health, Chemistry, Pharmacology and Chemical Biology, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY 10027, USA; Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health and Departments of Environmental Health, Chemistry, Pharmacology and Chemical Biology, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Wei Gu
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
28
|
Shin D, Lee J, Roh JL. Pioneering the future of cancer therapy: Deciphering the p53-ferroptosis nexus for precision medicine. Cancer Lett 2024; 585:216645. [PMID: 38280477 DOI: 10.1016/j.canlet.2024.216645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/21/2023] [Accepted: 01/11/2024] [Indexed: 01/29/2024]
Abstract
The TP53 gene, encoding the p53 protein, has been a focal point of research since its 1979 discovery, playing a crucial role in tumor suppression. Ferroptosis, a distinct form of cell death characterized by lipid peroxide accumulation, has gained prominence since its recognition in 2012. Recent studies have unveiled an intriguing connection between p53 and ferroptosis, with implications for cancer therapy. Recent research underscores p53 as a novel target for cancer therapy, influencing key metabolic processes in ferroptosis. Notably, p53 represses the expression of the cystine-glutamate antiporter SLC7A11, supporting p53-mediated tumor growth suppression. Furthermore, under metabolic stress, p53 mitigates ferroptosis sensitivity, aiding cancer cells in coping and delaying cell death. This dynamic interplay between p53 and ferroptosis has far-reaching implications for various diseases, particularly cancer. This review provides a comprehensive overview of ferroptosis in cancer cells, elucidating p53's role in regulating ferroptosis, and explores the potential of targeting p53 to induce ferroptosis for cancer therapy. Understanding this complex relationship between p53 and ferroptosis offers a promising avenue for developing innovative cancer treatments.
Collapse
Affiliation(s)
- Daiha Shin
- Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea.
| |
Collapse
|
29
|
Dibra D, Xiong S, Moyer SM, El-Naggar AK, Qi Y, Su X, Kong EK, Korkut A, Lozano G. Mutant p53 protects triple-negative breast adenocarcinomas from ferroptosis in vivo. SCIENCE ADVANCES 2024; 10:eadk1835. [PMID: 38354236 PMCID: PMC10866549 DOI: 10.1126/sciadv.adk1835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
The TP53 tumor suppressor gene is mutated early in most of the patients with triple-negative breast cancer (TNBC). The most frequent TP53 alterations are missense mutations that contribute to tumor aggressiveness. Here, we used an autochthonous somatic TNBC mouse model, in which mutant p53 can be toggled on and off genetically while leaving the tumor microenvironment intact and wild-type for p53 to identify physiological dependencies on mutant p53. In TNBCs that develop in this model, deletion of two different hotspot p53R172H and p53R245W mutants triggers ferroptosis in vivo, a cell death mechanism involving iron-dependent lipid peroxidation. Mutant p53 protects cells from ferroptosis inducers, and ferroptosis inhibitors reverse the effects of mutant p53 loss in vivo. Single-cell transcriptomic data revealed that mutant p53 protects cells from undergoing ferroptosis through NRF2-dependent regulation of Mgst3 and Prdx6, which encode two glutathione-dependent peroxidases that detoxify lipid peroxides. Thus, mutant p53 protects TNBCs from ferroptotic death.
Collapse
Affiliation(s)
- Denada Dibra
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shunbin Xiong
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sydney M. Moyer
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Genetics and Epigenetics Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Adel K. El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuan Qi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elisabeth K. Kong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Genetics and Epigenetics Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
30
|
Hu D, Zhang Z, Luo X, Li S, Jiang J, Zhang J, Wu Z, Wang Y, Sun M, Chen X, Zhang B, Xu X, Wang S, Xu S, Wang Y, Huang W, Xia L. Transcription factor BACH1 in cancer: roles, mechanisms, and prospects for targeted therapy. Biomark Res 2024; 12:21. [PMID: 38321558 PMCID: PMC10848553 DOI: 10.1186/s40364-024-00570-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Transcription factor BTB domain and CNC homology 1 (BACH1) belongs to the Cap 'n' Collar and basic region Leucine Zipper (CNC-bZIP) family. BACH1 is widely expressed in mammalian tissues, where it regulates epigenetic modifications, heme homeostasis, and oxidative stress. Additionally, it is involved in immune system development. More importantly, BACH1 is highly expressed in and plays a key role in numerous malignant tumors, affecting cellular metabolism, tumor invasion and metastasis, proliferation, different cell death pathways, drug resistance, and the tumor microenvironment. However, few articles systematically summarized the roles of BACH1 in cancer. This review aims to highlight the research status of BACH1 in malignant tumor behaviors, and summarize its role in immune regulation in cancer. Moreover, this review focuses on the potential of BACH1 as a novel therapeutic target and prognostic biomarker. Notably, the mechanisms underlying the roles of BACH1 in ferroptosis, oxidative stress and tumor microenvironment remain to be explored. BACH1 has a dual impact on cancer, which affects the accuracy and efficiency of targeted drug delivery. Finally, the promising directions of future BACH1 research are prospected. A systematical and clear understanding of BACH1 would undoubtedly take us one step closer to facilitating its translation from basic research into the clinic.
Collapse
Affiliation(s)
- Dian Hu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Zerui Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xiangyuan Luo
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Junqing Jiang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Zhangfan Wu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yijun Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xiaoping Chen
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China
| | - Bixiang Zhang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China
| | - Xiao Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Westlake university school of medicine, Hangzhou, 310006, China
| | - Shengjun Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
31
|
Wang Y, Wang S, Niu Y, Ma B, Li J. Data Mining Suggests That CXCL14 Gene Silencing in Colon Cancer Is Due to Promoter Methylation. Int J Mol Sci 2023; 24:16027. [PMID: 38003215 PMCID: PMC10671198 DOI: 10.3390/ijms242216027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
CXCL14 is one of the most evolutionarily conserved members of the chemokine family and is constitutionally expressed in multiple organs, suggesting that it is involved in the homeostasis maintenance of the system. CXCL14 is highly expressed in colon epithelial cells and shows obvious gene silencing in clinical colon cancer samples, suggesting that its silencing is related to the immune escape of cancer cells. In this paper, we analyzed the expression profiles of multiple human clinical colon cancer datasets and mouse colon cancer models to reveal the variation trend of CXCL14 expression during colitis, colon polyps, primary colon cancer, and liver metastases. The relationship between CXCL14 gene silencing and promoter hypermethylation was revealed through the colorectal carcinoma methylation database. The results suggest that CXCL14 is a tumor suppressor gene in colorectal carcinoma which is activated first and then silenced during the process of tumor occurrence and deterioration. Promoter hypermethylation is the main cause of CXCL14 silencing. The methylation level of CXCL14 is correlated with the anatomic site of tumor occurrence, positively correlated with patient age, and associated with prognosis. Reversing the hypermethylation of CXCL14 may be an epigenetic therapy for colon cancer.
Collapse
Affiliation(s)
| | | | | | - Buyong Ma
- Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.W.); (S.W.); (Y.N.)
| | - Jingjing Li
- Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.W.); (S.W.); (Y.N.)
| |
Collapse
|
32
|
Feng S, Tang D, Wang Y, Li X, Bao H, Tang C, Dong X, Li X, Yang Q, Yan Y, Yin Z, Shang T, Zheng K, Huang X, Wei Z, Wang K, Qi S. The mechanism of ferroptosis and its related diseases. MOLECULAR BIOMEDICINE 2023; 4:33. [PMID: 37840106 PMCID: PMC10577123 DOI: 10.1186/s43556-023-00142-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023] Open
Abstract
Ferroptosis, a regulated form of cellular death characterized by the iron-mediated accumulation of lipid peroxides, provides a novel avenue for delving into the intersection of cellular metabolism, oxidative stress, and disease pathology. We have witnessed a mounting fascination with ferroptosis, attributed to its pivotal roles across diverse physiological and pathological conditions including developmental processes, metabolic dynamics, oncogenic pathways, neurodegenerative cascades, and traumatic tissue injuries. By unraveling the intricate underpinnings of the molecular machinery, pivotal contributors, intricate signaling conduits, and regulatory networks governing ferroptosis, researchers aim to bridge the gap between the intricacies of this unique mode of cellular death and its multifaceted implications for health and disease. In light of the rapidly advancing landscape of ferroptosis research, we present a comprehensive review aiming at the extensive implications of ferroptosis in the origins and progress of human diseases. This review concludes with a careful analysis of potential treatment approaches carefully designed to either inhibit or promote ferroptosis. Additionally, we have succinctly summarized the potential therapeutic targets and compounds that hold promise in targeting ferroptosis within various diseases. This pivotal facet underscores the burgeoning possibilities for manipulating ferroptosis as a therapeutic strategy. In summary, this review enriched the insights of both investigators and practitioners, while fostering an elevated comprehension of ferroptosis and its latent translational utilities. By revealing the basic processes and investigating treatment possibilities, this review provides a crucial resource for scientists and medical practitioners, aiding in a deep understanding of ferroptosis and its effects in various disease situations.
Collapse
Affiliation(s)
- Shijian Feng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dan Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yichang Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiang Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Bao
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengbing Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiuju Dong
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinna Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinxue Yang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun Yan
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhijie Yin
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tiantian Shang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Kaixuan Zheng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaofang Huang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zuheng Wei
- Chengdu Jinjiang Jiaxiang Foreign Languages High School, Chengdu, People's Republic of China
| | - Kunjie Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Shiqian Qi
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
33
|
Irikura R, Nishizawa H, Nakajima K, Yamanaka M, Chen G, Tanaka K, Onodera M, Matsumoto M, Igarashi K. Ferroptosis model system by the re-expression of BACH1. J Biochem 2023; 174:239-252. [PMID: 37094356 DOI: 10.1093/jb/mvad036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 04/26/2023] Open
Abstract
Ferroptosis is a regulated cell death induced by iron-dependent lipid peroxidation. The heme-responsive transcription factor BTB and CNC homology 1 (BACH1) promotes ferroptosis by repressing the transcription of genes involved in glutathione (GSH) synthesis and intracellular labile iron metabolism, which are key regulatory pathways in ferroptosis. We found that BACH1 re-expression in Bach1-/- immortalized mouse embryonic fibroblasts (iMEFs) can induce ferroptosis upon 2-mercaptoethanol removal, without any ferroptosis inducers. In these iMEFs, GSH synthesis was reduced, and intracellular labile iron levels were increased upon BACH1 re-expression. We used this system to investigate whether the major ferroptosis regulators glutathione peroxidase 4 (Gpx4) and apoptosis-inducing factor mitochondria-associated 2 (Aifm2), the gene for ferroptosis suppressor protein 1, are target genes of BACH1. Neither Gpx4 nor Aifm2 was regulated by BACH1 in the iMEFs. However, we found that BACH1 represses AIFM2 transcription in human pancreatic cancer cells. These results suggest that the ferroptosis regulators targeted by BACH1 may vary across different cell types and animal species. Furthermore, we confirmed that the ferroptosis induced by BACH1 re-expression exhibited a propagating effect. BACH1 re-expression represents a new strategy for inducing ferroptosis after GPX4 or system Xc- suppression and is expected to contribute to future ferroptosis research.
Collapse
Key Words
- BACH1 Abbreviations: AIFM2, apoptosis-inducing factor mitochondria-associated 2; ANOVA, analysis of variance; BACH1, BTB and CNC homology 1; Bach1−/− mice, Bach1 knockout mice; BTB, Broad complex, Tramtrack, Bric-a-brac domain; bZIP, basic leucine zipper; ChIP-seq, chromatin immunoprecipitation sequencing; CNC, Cap‘n’Collar region; DAPI, 4′,6-diamidino-2-phenylindole; DFX, deferasirox; DMSO, dimethyl sulfoxide; EMT, epithelial–mesenchymal transition; Ferr-1, ferrostatin-1; FINs, ferroptosis inducers; FSP1, Ferroptosis suppressor protein 1; Fth1, ferritin heavy chain 1; Ftl, ferritin light chain; GCL, glutamate-cysteine ligase; Gclc, GCL catalytic subunit; Gclm, GCL modifier subunit; GEO, Gene Expression Omnibus; GPX4, glutathione peroxidase 4; GSH, glutathione; HO-1 (Hmox1), heme oxygenase 1; iMEFs, immortalized MEFs; KuO, Kusabira Orange; MAFK, musculoaponeurotic fibrosarcoma oncogene homolog bZIP transcription factor K; mBACH1, Bach1 gene of Mus musculus; 2-ME, 2-mercaptoethanol; MEFs, mouse embryonic fibroblasts; NRF2, nuclear factor-erythroid 2-related factor 2; NSA, necrosulfonamide; PDAC, pancreatic ductal adenocarcinoma; PI, Propidium iodide; Ptgs2, prostaglandin-endoperoxide synthase 2; RSL3, (1S,3R)-RSL3; Slc40a1, solute carrier family 40 member 1; Slc7a11, solute carrier family 7 member 11; TFRC, transferrin receptor 1; Z-VAD.FMK, Benzyloxycarbonyl-Val-Ala-Asp (OMe) fluoromethylketone
- extracellular signal
- ferroptosis
- fibroblasts
- transcription
Collapse
Affiliation(s)
- Riko Irikura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kazuma Nakajima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Gladstone Institute of Neurological Disease, Gladstone Institutes, 1650 Owens Street, San Francisco, CA 94158, USA
| | - Guan Chen
- Department of Molecular Oncology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Masafumi Onodera
- Gene & Cell Therapy Promotion Center, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|