1
|
He Y, Zhao G, Ouyang X, Wang S, Chen Y, Li C, He Y, Gao J, Han S, Zhao J, Wang J, Wang C. Creatine-mediated ferroptosis inhibition is involved in the intestinal radioprotection of daytime-restricted feeding. Gut Microbes 2025; 17:2489072. [PMID: 40205678 PMCID: PMC11988229 DOI: 10.1080/19490976.2025.2489072] [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: 10/26/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Ionizing radiation-induced intestinal injury (IRIII) is a catastrophic disease lack of sufficient medical countermeasures currently. Regulation of the gut microbiota through dietary adjustments is a potential strategy to mitigate IRIII. Time-restricted feeding (TRF) is an emerging behavioral nutrition intervention with pleiotropic health benefits. Whether this dietary pattern influences the pathogenesis of IRIII remains vague. We evaluated the impact of TRF on intestinal radiosensitivity in this study and discovered that only daytime TRF (DTRF), not nighttime TRF, could ameliorate intestinal damage in mice that received a high dose of IR. Faecal metagenomic and metabolomic studies revealed that the intestinal creatine level was increased by approximate 9 times by DTRF, to which the Bifidobacterium pseudolongum enrichment contribute. Further investigations showed that creatine could activate the energy sensor AMP-activated protein kinase in irradiated enterocytes and induce phosphorylation of acetyl-CoA carboxylase, resulting in reduced production of polyunsaturated fatty acids and reduced ferroptosis after IR. The administration of creatine mitigated IRIII and reduced bacteremia and proinflammatory responses. Blockade of creatine import compromised the ferroptosis inhibition and mitigation of DTRF on IRIII. Our study demonstrates a radioprotective dietary mode that can reshape the gut microbiota and increase intestinal creatine, which can suppress IR-induced ferroptosis, thereby providing effective countermeasures for IRIII prevention.
Collapse
Affiliation(s)
- Yingjuan He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Gaomei Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Xue Ouyang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Shaobo Wang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yin Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Chenwenya Li
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Yongwu He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jining Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Songling Han
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Liang W, Guo H, Li L, Tan W, Liu J, Hu X, Wang Y, Zhou S. Ferroptosis: a new target for depression prevention and treatment. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02912-4. [PMID: 40317298 DOI: 10.1007/s00702-025-02912-4] [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: 11/19/2024] [Accepted: 03/06/2025] [Indexed: 05/07/2025]
Abstract
Depression, a significant mental health issue, is one of the diseases with the highest disability rates worldwide. The exact etiology of depression remains undetermined, complicating the development of treatment strategies targeting specific mechanisms, and there is currently no effective cure. In this context, ferroptosis may represent a breakthrough in the understanding of depression. Ferroptosis is primarily associated with iron accumulation and lipid peroxidation, and recent studies have revealed its potential association with depression. Clinical evidence suggests that ferroptosis may influence the development and function of the hippocampus through interactions with neuroinflammation. Activated microglia, astrocytes, and neurons are involved in ferroptosis. This review summarizes recent findings on how ferroptosis contributes to depression, including glutathione peroxidase 4 (GPX4), nuclear factor-erythroid 2-related factor 2 (Nrf2), phase separation, and neuroinflammatory pathways, allowing the proposal of some new hypotheses. We hope that exploring the role of ferroptosis in the mechanism of depression will offer a new perspective on the complex biological basis of depression and provide theoretical support for the development of new therapeutic methods.
Collapse
Affiliation(s)
- Wenxuan Liang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China
- Basic Medical College, Guilin Medical College, Guilin, 541199, China
| | - Haowei Guo
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China
- Basic Medical College, Guilin Medical College, Guilin, 541199, China
| | - Luyao Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China
- Basic Medical College, Guilin Medical College, Guilin, 541199, China
| | - Wupeng Tan
- Department of Gynaecology, Maternal and Child Health Hospital of Hengyang, Hengyang, 421001, China
| | - Jianfeng Liu
- Department of Pediatrics, Second Affiliated Hospital of South China University, Hengyang, 421001, China
| | - Xiaoli Hu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China
- Basic Medical College, Guilin Medical College, Guilin, 541199, China
| | - Yuchu Wang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China.
- Basic Medical College, Guilin Medical College, Guilin, 541199, China.
| | - Shouhong Zhou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China.
- Basic Medical College, Guilin Medical College, Guilin, 541199, China.
| |
Collapse
|
3
|
Chen F, Chen L. CRISPR/Cas-mediated macromolecular DNA methylation editing: Precision targeting of DNA methyltransferases in cancer therapy. Int J Biol Macromol 2025; 308:142401. [PMID: 40132699 DOI: 10.1016/j.ijbiomac.2025.142401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Epigenetic modifications, particularly DNA methylation, play a pivotal role in gene regulation, influencing tumor suppressor silencing and oncogene activation in cancer. DNA methyltransferases (DNMTs), Ten-eleven translocation (TET) enzymes, and associated chromatin regulators are key biological macromolecules that mediate these epigenetic processes. Targeting aberrant DNA methylation holds great promise for cancer therapy, but traditional approaches lack precision and specificity. CRISPR/Cas-based epigenetic editing has emerged as a transformative tool for macromolecular DNA methylation reprogramming, offering targeted modifications without altering the genetic sequence. This review explores the role of DNMTs, TET enzymes, and chromatin-associated proteins in cancer epigenetics and discusses how CRISPR/dCas9 fused with DNMT3A or TET1 enables locus-specific DNA methylation editing. We highlight recent advances, including dCas9-DNMT3A for precise hypermethylation and dCas9-TET1 for targeted demethylation, and discuss their applications in reactivating tumor suppressor genes or silencing oncogenic pathways. Novel epigenetic editing systems, such as SunTag-based amplification, KRAB-MeCP2 repression, further enhance targeting efficiency and therapeutic potential. CRISPR/Cas-mediated macromolecular epigenetic editing represents a paradigm shift in cancer therapy, providing unprecedented control over DNA methylation and chromatin regulation. Despite challenges such as tumor heterogeneity and off-target effects, integrating CRISPR-based methylation reprogramming with precision oncology holds immense promise for future clinical applications.
Collapse
Affiliation(s)
- Feng Chen
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu 215500, Jiangsu Province, China.
| | - Lu Chen
- Pharma Technology A/S, Åshøjvej 24, 4600, Køge, Denmark.
| |
Collapse
|
4
|
Zheng J, Conrad M. Ferroptosis: when metabolism meets cell death. Physiol Rev 2025; 105:651-706. [PMID: 39661331 DOI: 10.1152/physrev.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
We present here a comprehensive update on recent advancements in the field of ferroptosis, with a particular emphasis on its metabolic underpinnings and physiological impacts. After briefly introducing landmark studies that have helped to shape the concept of ferroptosis as a distinct form of cell death, we critically evaluate the key metabolic determinants involved in its regulation. These include the metabolism of essential trace elements such as selenium and iron; amino acids such as cyst(e)ine, methionine, glutamine/glutamate, and tryptophan; and carbohydrates, covering glycolysis, the citric acid cycle, the electron transport chain, and the pentose phosphate pathway. We also delve into the mevalonate pathway and subsequent cholesterol biosynthesis, including intermediate metabolites like dimethylallyl pyrophosphate, squalene, coenzyme Q (CoQ), vitamin K, and 7-dehydrocholesterol, as well as fatty acid and phospholipid metabolism, including the biosynthesis and remodeling of ester and ether phospholipids and lipid peroxidation. Next, we highlight major ferroptosis surveillance systems, specifically the cyst(e)ine/glutathione/glutathione peroxidase 4 axis, the NAD(P)H/ferroptosis suppressor protein 1/CoQ/vitamin K system, and the guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin/dihydrofolate reductase axis. We also discuss other potential anti- and proferroptotic systems, including glutathione S-transferase P1, peroxiredoxin 6, dihydroorotate dehydrogenase, glycerol-3-phosphate dehydrogenase 2, vitamin K epoxide reductase complex subunit 1 like 1, nitric oxide, and acyl-CoA synthetase long-chain family member 4. Finally, we explore ferroptosis's physiological roles in aging, tumor suppression, and infection control, its pathological implications in tissue ischemia-reperfusion injury and neurodegeneration, and its potential therapeutic applications in cancer treatment. Existing drugs and compounds that may regulate ferroptosis in vivo are enumerated.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, Garching, Germany
| |
Collapse
|
5
|
Zhang C, Tian C, Zhu R, Chen C, Jin C, Wang X, Sun L, Peng W, Ji D, Zhang Y, Sun Y. CircSATB1 Promotes Colorectal Cancer Liver Metastasis through Facilitating FKBP8 Degradation via RNF25-Mediated Ubiquitination. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406962. [PMID: 39921520 PMCID: PMC11967755 DOI: 10.1002/advs.202406962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/14/2024] [Indexed: 02/10/2025]
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide and liver metastasis is the leading reason for its mortality. Circular RNAs (circRNAs) are conclusively associated with the progression of various cancers, rendering the exploration of its specific mechanisms in colorectal cancer liver metastasis(CRLM) highly valuable. Combined with GEO (Gene Expression Omnibus) databases and clinical data in our center, we found that high expression of circSATB1 is closely related to the progression of CRLM. Functionally, circSATB1 could significantly promote the metastatic ability of CRC cells in vitro and in vivo. Mechanistically, circSATB1 facilitated the RNF25-mediated ubiquitylation and degradation of FKBP8, releasing its inhibitory effects on mTOR signaling. In this process, circSATB1 acted as a scaffold for RNF25-FKBP8 complexes. Additionally, circSATB1 could be packaged in exosomes and secreted from the CRC primary tumors into plasma. In conclusion, this study uncovered a new circSATB1 that acts as a potent promoter of CRLM and offers novel insights into the precision therapeutic strategies for CRLM.
Collapse
Affiliation(s)
- Chuan Zhang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Chuanxin Tian
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Renzhong Zhu
- Institute of Translational Medicine, Medical CollegeYangzhou UniversityYangzhou225000China
| | - Chen Chen
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Chi Jin
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Xiaowei Wang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Lejia Sun
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Wen Peng
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Dongjian Ji
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Yue Zhang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| | - Yueming Sun
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityColorectal Institute of Nanjing Medical UniversityNanjing210000China
| |
Collapse
|
6
|
Zhou P, Peng X, Zhang K, Cheng J, Tang M, Shen L, Zhou Q, Li D, Yang L. HAT1/HDAC2 mediated ACSL4 acetylation confers radiosensitivity by inducing ferroptosis in nasopharyngeal carcinoma. Cell Death Dis 2025; 16:160. [PMID: 40050614 PMCID: PMC11885570 DOI: 10.1038/s41419-025-07477-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 02/01/2025] [Accepted: 02/24/2025] [Indexed: 03/09/2025]
Abstract
Protein acetylation modification plays important roles in various aspects of tumor progression. Ferroptosis driven by lethal lipid peroxidation is closely related to tumor development. Targeting ferroptosis has become a promising strategy. However, the crosstalk between protein acetylation and ferroptosis remains unclear. In present study, we found that the acetylation of acyl-CoA synthase long-chain family member 4 (ACSL4) enhances its protein stability and a double-edged sword regulation in nasopharyngeal carcinoma (NPC). On the one hand, ACSL4 could promote the malignant progress of tumors; on the other hand, it enhanced radiosensitivity by endowing NPC cells with ferroptosis-sensitive properties in vitro and in vivo. Mechanistically, histone acetyltransferase 1 (HAT1) directly promotes the acetylation of ACSL4 at lysine 383, and deacetylase sirtuin 3 (SIRT3) mediates the deacetylation of ACSL4. Meanwhile, another deacetylase histone deacetylase 2 (HDAC2) enhances ACSL4 acetylation through inhibiting the transcription of SIRT3. Acetylation of ACSL4 inhibits F-box protein 10 (FBXO10)-mediated K48-linked ubiquitination, resulting in enhanced protein stability of ACSL4. This study reveals the novel regulatory mechanism of ferroptosis-related protein from the perspective of protein acetylation, and provides a novel method for the radiosensitivity of NPC.
Collapse
Affiliation(s)
- Peijun Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xingzhi Peng
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, China
| | - Kun Zhang
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jin Cheng
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, China
| | - Min Tang
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, China
| | - Lin Shen
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qin Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Dan Li
- Department of Life Science, College of Biology, Hunan University, Changsha, China.
| | - Lifang Yang
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, China.
| |
Collapse
|
7
|
Dhang S, Mondal A, Das C, Roy S. Metformin inhibits the histone methyltransferase CARM1 and attenuates H3 histone methylation during gluconeogenesis. J Biol Chem 2025; 301:108271. [PMID: 39922487 PMCID: PMC11910104 DOI: 10.1016/j.jbc.2025.108271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/18/2025] [Accepted: 01/30/2025] [Indexed: 02/10/2025] Open
Abstract
Hyperglycemia is a hallmark of metabolic disorders, yet the precise mechanisms linking epigenetic regulation to glucose metabolism remain underexplored. Coactivator-associated arginine methyltransferase 1 (CARM1), a type I histone methyltransferase, promotes transcriptional activation through the methylation of histone H3 at arginine residues H3R17 and H3R26. Here, we identify a novel mechanism by which metformin, a widely prescribed antidiabetic drug, inhibits CARM1 activity. Using biochemical and biophysical assays, we show that metformin binds to the substrate-binding site of CARM1, reducing histone H3 methylation levels in CARM1-overexpressing hepatic cells and liver tissues from metformin-fed mice. This epigenetic modulation suppresses the expression of gluconeogenic enzymes (G6Pase, FBPase, and PCK1), thereby reversing CARM1-induced glycolytic suppression and regulating gluconeogenesis. Importantly, metformin does not alter CARM1 protein levels and its recruitment to gluconeogenic gene promoters but diminishes H3R17me2a marks at these loci. Our findings reveal a previously unrecognized epigenetic mechanism of metformin action, offering new therapeutic insights for hyperglycemia management.
Collapse
Affiliation(s)
- Sinjini Dhang
- Structural Biology and Bio-Informatics Division, Council of Scientific & Industrial Research-Indian Institute of Chemical Biology, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India
| | - Siddhartha Roy
- Structural Biology and Bio-Informatics Division, Council of Scientific & Industrial Research-Indian Institute of Chemical Biology, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
8
|
Li S, Yuan H, Li L, Li Q, Lin P, Li K. Oxidative Stress and Reprogramming of Lipid Metabolism in Cancers. Antioxidants (Basel) 2025; 14:201. [PMID: 40002387 PMCID: PMC11851681 DOI: 10.3390/antiox14020201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Oxidative stress is a common event involved in cancer pathophysiology, frequently accompanied by unique lipid metabolic reprogramming phenomena. Oxidative stress is caused mainly by an imbalance between the production of reactive oxygen species (ROS) and the antioxidant system in cancer cells. Emerging evidence has reported that oxidative stress regulates the expression and activity of lipid metabolism-related enzymes, leading to the alteration of cellular lipid metabolism; this involves a significant increase in fatty acid synthesis and a shift in the way in which lipids are taken up and utilized. The dysregulation of lipid metabolism provides abundant intermediates to synthesize biological macromolecules for the rapid proliferation of cancer cells; moreover, it contributes to the maintenance of intracellular redox homeostasis by producing a variety of reducing agents. Moreover, lipid derivatives and metabolites play critical roles in signal transduction within cancer cells and in the tumor microenvironment that evades immune destruction and facilitates tumor invasion and metastasis. These findings suggest a close relationship between oxidative stress and lipid metabolism during the malignant progression of cancers. This review focuses on the crosstalk between the redox system and lipid metabolic reprogramming, which provides an in-depth insight into the modulation of ROS on lipid metabolic reprogramming in cancers and discusses potential strategies for targeting lipid metabolism for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Ping Lin
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (S.L.); (H.Y.); (L.L.); (Q.L.)
| | - Kai Li
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (S.L.); (H.Y.); (L.L.); (Q.L.)
| |
Collapse
|
9
|
Huang J, Qiao B, Yuan Y, Xie Y, Xia X, Li F, Wang L. PRMT3 and CARM1: Emerging Epigenetic Targets in Cancer. J Cell Mol Med 2025; 29:e70386. [PMID: 39964832 PMCID: PMC11834966 DOI: 10.1111/jcmm.70386] [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: 03/11/2024] [Revised: 01/01/2025] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
The family of protein arginine methyltransferases (PRMTs) occupies an important position in biology, especially during the initiation and development of cancer. PRMT3 and CARM1(also known as PRMT4), being type I protein arginine methyltransferases, are key in controlling tumour progression by catalysing the mono-methylation and asymmetric di-methylation of both histone and non-histone substrates. This paper reviews the functions and potential therapeutic target value of PRMT3 and CARM1 in a variety of cancers. Studies have identified abnormal expressions of PRMT3 and CARM1 in several malignancies, closely linked to cancer progression, advancement, and resistance to treatment. Such as hepatocellular carcinoma, colorectal cancer, ovarian cancer, and endometrial cancer. These findings offer new strategies and directions for cancer treatment, especially in enhancing the effectiveness of conventional treatment methods.
Collapse
Affiliation(s)
- Jiezuo Huang
- College of Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Beining Qiao
- College of Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Yixin Yuan
- Xiangya College of Public HealthCentral South UniversityChangshaChina
| | - Yuxuan Xie
- Hunan Normal University School of MedicineChangshaChina
| | - Xiaomeng Xia
- Department of Gynaecology and Obstetrics, Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Fenghe Li
- Department of Gynaecology and Obstetrics, Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Lei Wang
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical ScienceCentral South UniversityChangshaChina
| |
Collapse
|
10
|
Liu W, Zhu Y, Ye W, Xiong J, Wang H, Gao Y, Huang S, Zhang Y, Zhou X, Zhou X, Ge X, Cai W, Zheng X. Redox regulation of TRIM28 facilitates neuronal ferroptosis by promoting SUMOylation and inhibiting OPTN-selective autophagic degradation of ACSL4. Cell Death Differ 2025:10.1038/s41418-025-01452-4. [PMID: 39875520 DOI: 10.1038/s41418-025-01452-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 01/30/2025] Open
Abstract
Ferroptosis is one of the cell death programs occurring after spinal cord injury (SCI) and is driven by iron-dependent phospholipid peroxidation. However, little is known about its underlying regulation mechanism. The present study demonstrated that lipid peroxidation was promoted in patients with SCI. Neurons affected by ferroptosis following SCI had a high expression of ferroptotic protein ACSL4. The E3 SUMOylase TRIM28 promoted neuronal ferroptosis by enhancing ACSL4 expression. Genetic deletion of Trim28 significantly attenuated neuronal ferroptosis and improved mouse hindlimb motor function following SCI. In contrast, mice with Trim28 overexpression demonstrated poor neurological function after SCI, which was attenuated by ferroptosis inhibitor Liproxstatin-1. Mechanistically, TRIM28 bound to ACSL4, promoted SUMO3 modification at lysine (K) 532, and inhibited K63-linked ACSL4 ubiquitination, thereby suppressing OPTN-dependent autophagic degradation. Additionally, SENP3 was identified as the deSUMOylation enzyme that can reverse this process and compete with TRIM28, which was transcriptionally upregulated due to excessive oxidative stress. These data unveiled a mechanism by which TRIM28-mediated SUMOylation regulated neuronal ACSL4 levels and ferroptosis, identified interactions and correlations involved in ACSL4 SUMOylation, ubiquitination, and autophagic degradation, and discovered a positive feedback loop where oxidative stress transcriptionally upregulated Trim28, and conversely TRIM28 promoted ferroptosis and oxidative stress. Notably, screening of the FDA-approved drug library revealed that pharmacological TRIM28/ACSL4 axis interventions with Rutin hydrate inhibited neuronal ferroptosis and improved hindlimb motor function in mice after SCI, thus providing a promising therapeutic strategy for its treatment.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopedics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai, 200433, China.
| | - Yufeng Zhu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wu Ye
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Junjun Xiong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Haofan Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yu Gao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Shixue Huang
- Department of Orthopedics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Yinuo Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Xin Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Xuhui Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai, 200433, China.
- Translational Research Centre of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Xuhui Ge
- Department of Orthopedics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai, 200433, China.
| | - Weihua Cai
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Xingdong Zheng
- Translational Research Centre of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- Total Quality Management Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
11
|
Yang Y, Yu S, Liu W, Zhuo Y, Qu C, Zeng Y. Ferroptosis-related signaling pathways in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:1. [PMID: 39935430 PMCID: PMC11813627 DOI: 10.20517/cdr.2024.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
Ferroptosis is an iron-dependent form of programmed cell death induced by lipid peroxidation. This process is regulated by signaling pathways associated with redox balance, iron metabolism, and lipid metabolism. Cancer cells' increased iron demand makes them especially susceptible to ferroptosis, significantly influencing cancer development, therapeutic response, and metastasis. Recent findings indicate that cancer cells can evade ferroptosis by downregulating key signaling pathways related to this process, contributing to drug resistance. This underscores the possibility of modulating ferroptosis as an approach to counteract drug resistance and enhance therapeutic efficacy. This review outlines the signaling pathways involved in ferroptosis and their interactions with cancer-related signaling pathways. We also highlight the current understanding of ferroptosis in cancer drug resistance, offering insights into how targeting ferroptosis can provide novel therapeutic approaches for drug-resistant cancers. Finally, we explore the potential of ferroptosis-inducing compounds and examine the challenges and opportunities for drug development in this evolving field.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Simin Yu
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wanyao Liu
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Yi Zhuo
- First Clinical Department of Changsha Medical University, Changsha 410219, Hunan, China
| | - Chunrun Qu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
12
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
13
|
Wang Y, Cao X, Yang C, Fan J, Zhang X, Wu X, Guo W, Sun S, Liu M, Zhang L, Li T. Ferroptosis and immunosenescence in colorectal cancer. Semin Cancer Biol 2024; 106-107:156-165. [PMID: 39419366 DOI: 10.1016/j.semcancer.2024.10.003] [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: 08/08/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
Colorectal cancer (CRC), ranked as the globe's third leading malignancy. Despite advancements in therapeutic approaches, the mortality rate remains distressingly high for those afflicted with advanced stages of the disease. Ferroptosis is a programmed form of cell death. The ways of ferroptosis mainly include promoting the accumulation of cellular ROS and increasing the level of cellular Labile iron pool (LIP). Immunosenescence is characterized by a gradual deterioration of the immune system's ability to respond to pathogens and maintain surveillance against cancer cells. In CRC, this decline is exacerbated by the tumor microenvironment, which can suppress the immune response and promote tumor progression. This paper reviews the relationship between iron prolapse and immune senescence in colorectal cancer, focusing on the following aspects: firstly, the different pathways that induce iron prolapse in colorectal cancer; secondly, immune-immune senescence in colorectal cancer; and lastly, the interactions between immune senescence and iron prolapse in colorectal cancer, e.g., immune-immune senescent cells often exhibit increased oxidative stress, leading to the accumulation of ROS, and consequently to lipid peroxidation and induction of iron-induced cell death. At the same time, ferroptosis induces immune cell senescence as well as alterations in the immune microenvironment by promoting the death of damaged or diseased cells and leading to the inflammation usually associated with it. In conclusion, by exploring the potential targets of ferroptosis and immune senescence in colorectal cancer therapy, we hope to provide a reference for future research.
Collapse
Affiliation(s)
- Yao Wang
- Inpatient ward 8, General Surgery, Harbin Medical University Affiliated Second Hospital, Harbin 150000, China
| | - Xinran Cao
- Graduate School, Hebei North University, Zhangjiakou 075000, China
| | - Chunbaixue Yang
- Graduate School, Hebei North University, Zhangjiakou 075000, China
| | - Jianchun Fan
- Institute of Cancer, The First Affiliated Hospital of Hebei North University, Hebei 075000, China
| | - Xingmei Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China.
| | - Xueliang Wu
- Institute of Cancer, The First Affiliated Hospital of Hebei North University, Hebei 075000, China; Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou 075000, China.
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Shoutian Sun
- Department of Emergency, Zibo Central Hospital, Zibo 255024, China.
| | - Ming Liu
- General Surgery, Harbin Medical University Affiliated Fourth Hospital, Harbin 150000, China.
| | - Lifen Zhang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
14
|
Wang Y, Yan D, Liu J, Tang D, Chen X. Protein modification and degradation in ferroptosis. Redox Biol 2024; 75:103259. [PMID: 38955112 PMCID: PMC11267077 DOI: 10.1016/j.redox.2024.103259] [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: 03/08/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Ferroptosis is a form of iron-related oxidative cell death governed by an integrated redox system, encompassing pro-oxidative proteins and antioxidative proteins. These proteins undergo precise control through diverse post-translational modifications, including ubiquitination, phosphorylation, acetylation, O-GlcNAcylation, SUMOylation, methylation, N-myristoylation, palmitoylation, and oxidative modification. These modifications play pivotal roles in regulating protein stability, activity, localization, and interactions, ultimately influencing both the buildup of iron and lipid peroxidation. In mammalian cells, regulators of ferroptosis typically undergo degradation via two principal pathways: the ubiquitin-proteasome system, which handles the majority of protein degradation, and autophagy, primarily targeting long-lived or aggregated proteins. This comprehensive review aims to summarize recent advances in the post-translational modification and degradation of proteins linked to ferroptosis. It also discusses strategies for modulating ferroptosis through protein modification and degradation systems, providing new insights into potential therapeutic applications for both cancer and non-neoplastic diseases.
Collapse
Affiliation(s)
- Yuan Wang
- Key Laboratory of Biological Targeting Diagnosis, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ding Yan
- Key Laboratory of Biological Targeting Diagnosis, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jinbao Liu
- Key Laboratory of Biological Targeting Diagnosis, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China; Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 511436, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, 75390, USA.
| | - Xin Chen
- Key Laboratory of Biological Targeting Diagnosis, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
15
|
Li Y, Cheng X. Enhancing Colorectal Cancer Immunotherapy: The Pivotal Role of Ferroptosis in Modulating the Tumor Microenvironment. Int J Mol Sci 2024; 25:9141. [PMID: 39273090 PMCID: PMC11395055 DOI: 10.3390/ijms25179141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant challenge in oncology, with increasing incidence and mortality rates worldwide, particularly among younger adults. Despite advancements in treatment modalities, the urgent need for more effective therapies persists. Immunotherapy has emerged as a beacon of hope, offering the potential for improved outcomes and quality of life. This review delves into the critical interplay between ferroptosis, an iron-dependent form of regulated cell death, and immunotherapy within the CRC context. Ferroptosis's influence extends beyond tumor cell fate, reshaping the tumor microenvironment (TME) to enhance immunotherapy's efficacy. Investigations into Ferroptosis-related Genes (OFRGs) reveal their pivotal role in modulating immune cell infiltration and TME composition, closely correlating with tumor responsiveness to immunotherapy. The integration of ferroptosis inducers with immunotherapeutic strategies, particularly through novel approaches like ferrotherapy and targeted co-delivery systems, showcases promising avenues for augmenting treatment efficacy. Furthermore, the expression patterns of OFRGs offer novel prognostic tools, potentially guiding personalized and precision therapy in CRC. This review underscores the emerging paradigm of leveraging ferroptosis to bolster immunotherapy's impact, highlighting the need for further research to translate these insights into clinical advancements. Through a deeper understanding of the ferroptosis-immunotherapy nexus, new therapeutic strategies can be developed, promising enhanced efficacy and broader applicability in CRC treatment, ultimately improving patient outcomes and quality of life in the face of this formidable disease.
Collapse
Affiliation(s)
- Yanqing Li
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Xiaofei Cheng
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
16
|
Wang D, Kang X, Zhang L, Guo Y, Zhang Z, Ren H, Yuan G. TRIB2-Mediated Modulation of AMPK Promotes Hepatic Insulin Resistance. Diabetes 2024; 73:1199-1214. [PMID: 38394623 DOI: 10.2337/db23-0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Abstract
Insulin resistance and its linked health complications are increasing in prevalence. Recent work has caused the role of Tribbles2 (TRIB2) in metabolism and cellular signaling to be increasingly appreciated, but its role in the progression of insulin resistance has not been elucidated. Here, we explore the functions of TRIB2 in modulating insulin resistance and the mechanism involved in insulin-resistant mice and palmitic acid-treated HepG2 cells. We demonstrate that whole-body knockout and hepatic-specific TRIB2 deficiency protect against diet-induced insulin resistance, inflammation, and endoplasmic reticulum stress. Accordingly, upregulation of TRIB2 in the liver aggravates these metabolic disturbances in high-fat diet-induced mice and ob/ob mice. Mechanistically, TRIB2 directly binds to the αγ-SBS domain of PRKAB through its pseudokinase domain, subsequently inhibiting the formation and activity of the AMPK complex. Moreover, the results of intervention against AMPK suggest that the effects of TRIB2 depend on AMPK. Our findings reveal that TRIB2 is a novel target for the treatment of insulin resistance and its associated metabolic complications and clarify the function of TRIB2 as a regulatory component of AMPK activity. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Dan Wang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xiaonan Kang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Lu Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yaoyao Guo
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ziyin Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Huihui Ren
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Clinical Research Center for Metabolic Disease, Wuhan, Hubei, People's Republic of China
| | - Gang Yuan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Clinical Research Center for Metabolic Disease, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
17
|
Adzavon KP, Zhao W, He X, Sheng W. Ferroptosis resistance in cancer cells: nanoparticles for combination therapy as a solution. Front Pharmacol 2024; 15:1416382. [PMID: 38962305 PMCID: PMC11219589 DOI: 10.3389/fphar.2024.1416382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/20/2024] [Indexed: 07/05/2024] Open
Abstract
Ferroptosis is a form of regulated cell death (RCD) characterized by iron-dependent lipid peroxidation. Ferroptosis is currently proposed as one of the most promising means of combating tumor resistance. Nevertheless, the problem of ferroptosis resistance in certain cancer cells has been identified. This review first, investigates the mechanisms of ferroptosis induction in cancer cells. Next, the problem of cancer cell resistance to ferroptosis, as well as the underlying mechanisms is discussed. Recently discovered ferroptosis-suppressing biomarkers have been described. The various types of nanoparticles that can induce ferroptosis are also discussed. Given the ability of nanoparticles to combine multiple agents, this review proposes nanoparticle-based ferroptosis cell death as a viable method of circumventing this resistance. This review suggests combining ferroptosis with other forms of cell death, such as apoptosis, cuproptosis and autophagy. It also suggests combining ferroptosis with immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Wang Sheng
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| |
Collapse
|
18
|
Wu Y, Shen Y. Research Progress on CARM1 and its Relationship with Colorectal Cancer. Cancer Invest 2024; 42:435-442. [PMID: 38813691 DOI: 10.1080/07357907.2024.2354798] [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: 04/03/2024] [Accepted: 05/09/2024] [Indexed: 05/31/2024]
Abstract
Coactivator-associated arginine methyltransferase 1 (CARM1) is significant as a key member of the PRMT family, crucial for regulating arginine methylation, and its association with colorectal cancer underscores its potential as a therapeutic target. Consequently, CARM1 inhibitors have emerged as potential therapeutic agents in cancer treatment and valuable chemical tools for cancer research. Despite steady progress in CARM1 inhibitor research, challenges persist in discovering effective, isoform-selective, cell-permeable, and in vivo-active CARM1 inhibitors for colorectal cancer. This review summarizes the research progress on CARM1 and its relationship with colorectal cancer, aiming to provide a theoretical basis for the radiotherapy of colorectal cancer.
Collapse
Affiliation(s)
- Yuchen Wu
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Yannan Shen
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
19
|
Chen R, Zhu S, Zhao R, Liu W, Jin L, Ren X, He H. Targeting ferroptosis as a potential strategy to overcome the resistance of cisplatin in oral squamous cell carcinoma. Front Pharmacol 2024; 15:1402514. [PMID: 38711989 PMCID: PMC11071065 DOI: 10.3389/fphar.2024.1402514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 03/29/2024] [Indexed: 05/08/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a crucial public health problem, accounting for approximately 2% of all cancers globally and 90% of oral malignancies over the world. Unfortunately, despite the achievements in surgery, radiotherapy, and chemotherapy techniques over the past decades, OSCC patients still low 5-year survival rate. Cisplatin, a platinum-containing drug, serves as one of the first-line chemotherapeutic agents of OSCC. However, the resistance to cisplatin significantly limits the clinical practice and is a crucial factor in tumor recurrence and metastasis after conventional treatments. Ferroptosis is an iron-based form of cell death, which is initiated by the intracellular accumulation of lipid peroxidation and reactive oxygen species (ROS). Interestingly, cisplatin-resistant OSCC cells exhibit lower level of ROS and lipid peroxidation compared to sensitive cells. The reduced ferroptosis in cisplatin resistance cells indicates the potential relationship between cisplatin resistance and ferroptosis, which is proved by recent studies showing that in colorectal cancer cells. However, the modulation pathway of ferroptosis reversing cisplatin resistance in OSCC cells still remains unclear. This article aims to concisely summarize the molecular mechanisms and evaluate the relationship between ferroptosis and cisplatin resistance OSCC cells, thereby providing novel strategies for overcoming cisplatin resistance and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Rongkun Chen
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Shuyu Zhu
- Department of Oral Implantology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Ruoyu Zhao
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Wang Liu
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Luxin Jin
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Xiaobin Ren
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Hongbing He
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| |
Collapse
|
20
|
Liu C, Li Y, Liu Z, Cao C, Lin M, Chen X, Yuan M, Fan Y, Gu X, Wang L, Yang F, Ye F, Jin J. Structure-based discovery of potent CARM1 inhibitors for colorectal cancer therapy. Eur J Med Chem 2024; 269:116288. [PMID: 38460270 DOI: 10.1016/j.ejmech.2024.116288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/11/2024]
Abstract
Coactivator-associated arginine methyltransferase 1 (CARM1) plays an important role in cell proliferation and gene expression, and is highly expressed in a variety of tumor tissues. Guided by our previous reported structure of DCPR049_12, we focused on designing and evaluating selective CARM1 inhibitors, resulting in the identification of compound 11f as a promising lead candidate. Compound 11f displayed potent inhibition of CARM1 (IC50 = 9 nM). Comprehensive evaluations, including in vitro metabolic stability assessments, molecular modelling, cellular studies, and in vivo anti-tumor studies, confirmed that it induced cancer cell apoptosis and specifically inhibited CARM1's methylation function. Notably, compound 11f displayed significant anti-proliferative effects on colorectal cancer cell lines, showcasing its potential for targeted therapies against CARM1-related diseases. This study provides valuable insights for the future development of specific and effective CARM1 inhibitors.
Collapse
Affiliation(s)
- Chenyu Liu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Yang Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Zhihao Liu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Chenxi Cao
- Department of Oncology, The Second Affiliated Hospital of Jiaxing University, No. 397, Huangcheng North Road, Jiaxing, 314000, China
| | - Min Lin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xin Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Mengting Yuan
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yaohua Fan
- Department of Oncology, The Second Affiliated Hospital of Jiaxing University, No. 397, Huangcheng North Road, Jiaxing, 314000, China
| | - Xiaodong Gu
- Department of Oncology, The Second Affiliated Hospital of Jiaxing University, No. 397, Huangcheng North Road, Jiaxing, 314000, China
| | - Lei Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Fan Yang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China.
| | - Fei Ye
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Jia Jin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|