1
|
Zhang S, Huang Q, Ji T, Li Q, Hu C. Copper homeostasis and copper-induced cell death in tumor immunity: implications for therapeutic strategies in cancer immunotherapy. Biomark Res 2024; 12:130. [PMID: 39482784 PMCID: PMC11529036 DOI: 10.1186/s40364-024-00677-8] [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: 07/10/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024] Open
Abstract
Copper is an important trace element for maintaining key biological functions such as cellular respiration, nerve conduction, and antioxidant defense. Maintaining copper homeostasis is critical for human health, and its imbalance has been linked to various diseases, especially cancer. Cuproptosis, a novel mechanism of copper-induced cell death, provides new therapeutic opportunities for metal ion regulation to interact with cell fate. This review provides insights into the complex mechanisms of copper metabolism, the molecular basis of cuproptosis, and its association with cancer development. We assess the role of cuproptosis-related genes (CRGs) associated with tumorigenesis, their importance as prognostic indicators and therapeutic targets, and the impact of copper homeostasis on the tumor microenvironment (TME) and immune response. Ultimately, this review highlights the complex interplay between copper, cuproptosis, and cancer immunotherapy.
Collapse
Affiliation(s)
- Suhang Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430030, China
| | - Qibo Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tuo Ji
- School of Medicine, New York Medical College, Valhalla, NY, 10595, USA
| | - Qilin Li
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430030, China.
| | - Chuanyu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430030, China.
| |
Collapse
|
2
|
He T, Kang J, Tang X, Wu Y, Hao L. Overexpression of MCL1 attenuates irritable bowel syndrome by regulating cuproptosis: Screening and validation. Biochem Biophys Res Commun 2024; 737:150926. [PMID: 39488085 DOI: 10.1016/j.bbrc.2024.150926] [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: 07/23/2024] [Revised: 09/30/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Irritable bowel syndrome (IBS) is a type of chronic bowel disorder with a poorly understood pathophysiology. Recently, the imbalance of copper has been reported to influence the progression of IBS, suggesting cuproptosis, a new type of copper-induced cell death, may play a role in IBS. This study found 17 cuproptosis-related differentially expressed genes in IBS through bioinformatic analysis. Six hub genes were identified after the protein-protein interaction network analysis, namely myeloid cell leukemia 1 (MCL1), epidermal growth factor receptor 2, cadherin-associated protein beta 1, solute carrier family 25 members 37, solute carrier family 39 members 14, and six transmembrane epithelial antigens of the prostate 3. We selected MCL1 for further verification. Human normal colon epithelial cell line (NCM460) was used to construct models of IBS or cuproptosis in vitro by lipopolysaccharide (LPS) or LPS combined with copper (II) chloride (CuCl2). We observed that overexpression of MCL1 promoted cell viability and proliferation ability, and inhibited the secretion of inflammatory factors and expression of Bax and caspase-3 of NCM460 cells treated with LPS or LPS combined with CuCl2. In addition, up-regulated MCL1 significantly suppressed the protein levels of ferredoxin 1 and lipoyl synthase, two key regulators of cuproptosis. In conclusion, our study demonstrates that cuproptosis is involved in IBS and identifies a cuproptosis-related gene, MCL1, that helps alleviate IBS by promoting cell growth, reducing inflammation, and suppressing cuproptosis, making it a promising therapeutic target in IBS.
Collapse
Affiliation(s)
- Taohong He
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China
| | - Jian Kang
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China; School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, No.37-39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610075, China
| | - Xiao Tang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Meishuguan Back Street, Dongcheng District, Beijing, 100010, China
| | - Yuqi Wu
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China; School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, No.37-39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610075, China
| | - Liangliang Hao
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China.
| |
Collapse
|
3
|
Li Y, Zhang H, Yang F, Zhu D, Chen S, Wang Z, Wei Z, Yang Z, Jia J, Zhang Y, Wang D, Ma M, Kang X. Mechanisms and therapeutic potential of disulphidptosis in cancer. Cell Prolif 2024:e13752. [PMID: 39354653 DOI: 10.1111/cpr.13752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
SLC7A11 plays a pivotal role in tumour development by facilitating cystine import to enhance glutathione synthesis and counteract oxidative stress. Disulphidptosis, an emerging form of cell death observed in cells with high expression of SLC7A11 under glucose deprivation, is regulated through reduction-oxidation reactions and disulphide bond formation. This process leads to contraction and collapse of the F-actin cytoskeleton from the plasma membrane, ultimately resulting in cellular demise. Compared to other forms of cell death, disulphidptosis exhibits distinctive characteristics and regulatory mechanisms. This mechanism provides novel insights and innovative strategies for cancer treatment while also inspiring potential therapeutic approaches for other diseases. Our review focuses on elucidating the molecular mechanism underlying disulphidptosis and its connection with the actin cytoskeleton, identifying alternative metabolic forms of cell death, as well as offering insights into disulphidptosis-based cancer therapy. A comprehensive understanding of disulphidptosis will contribute to our knowledge about fundamental cellular homeostasis and facilitate the development of groundbreaking therapies for disease treatment.
Collapse
Affiliation(s)
- Yanhu Li
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Haijun Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
- The Second People's Hospital of Gansu Province, Lanzhou, PR China
| | - Fengguang Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Daxue Zhu
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Shijie Chen
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhaoheng Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Ziyan Wei
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhili Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Jingwen Jia
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Yizhi Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Dongxin Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Mingdong Ma
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Xuewen Kang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| |
Collapse
|
4
|
Peng G, Huang Y, Xie G, Tang J. Exploring Copper's role in stroke: progress and treatment approaches. Front Pharmacol 2024; 15:1409317. [PMID: 39391696 PMCID: PMC11464477 DOI: 10.3389/fphar.2024.1409317] [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: 03/29/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Copper is an important mineral, and moderate copper is required to maintain physiological processes in nervous system including cerebral ischemia/reperfusion (I/R) injury. Over the past few decades, copper induced cell death, named cuprotosis, has attracted increasing attention. Several lines of evidence have confirmed cuprotosis exerts pivotal role in diverse of pathological processes, such as cancer, neurodegenerative diseases, and I/R injury. Therefore, an in-depth understanding of the interaction mechanism between copper-mediated cell death and I/R injury may reveal the significant alterations about cellular copper-mediated homeostasis in physiological and pathophysiological conditions, as well as therapeutic strategies deciphering copper-induced cell death in cerebral I/R injury.
Collapse
Affiliation(s)
- Gang Peng
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan, China
| | - Yongpan Huang
- School of Medicine, Changsha Social Work College, Changsha, Hunan, China
| | - Guangdi Xie
- Department of Neurology, Huitong People’s Hospital, Huitong, Hunan, China
| | - Jiayu Tang
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
5
|
Qin L, Cao X, Huang T, Liu Y, Li S. Identification of potential biomarkers of cuproptosis in cerebral ischemia. Front Nutr 2024; 11:1410431. [PMID: 39360273 PMCID: PMC11445069 DOI: 10.3389/fnut.2024.1410431] [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: 04/01/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Objective Cerebral ischemia can cause mild damage to local brain nerves due to hypoxia and even lead to irreversible damage due to neuronal cell death. However, the underlying pathogenesis of this phenomenon remains unclear. This study utilized bioinformatics to explore the role of cuproptosis in cerebral ischemic disease and its associated biomarkers. Method R software identified the overlap of cerebral ischemia and cuproptosis genes, analyzed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), and explored hub genes. Expressions and localizations of hub genes in brain tissue, cells, and immune cells were analyzed, along with predictions of protein structures, miRNAs, and transcription factors. A network was constructed depicting hub gene co-expression with miRNAs and interactions with transcription factors. Ferredoxin 1 (FDX1) expression was determined using qRT-PCR. Results Ten cuproptosis-related genes in cerebral ischemia were identified, with GO analysis revealing involvement in acetyl-CoA synthesis, metabolism, mitochondrial function, and iron-sulfur cluster binding. KEGG highlighted processes like the tricarboxylic acid cycle, pyruvate metabolism, and glycolysis/gluconeogenesis. Using the Human Protein Atlas, eight hub genes associated with cuproptosis were verified in brain tissues, hippocampus, and AF22 cells. Lipoyl(octanoyl) transferase 1 (LIPT1), was undetected, while others were found in mitochondria or both nucleus and mitochondria. These genes were differentially expressed in immune cells. FDX1, lipoic acid synthetase (LIAS), dihydrolipoamide S-acetyltransferase (DLAT), pyruvate dehydrogenase E1 component subunit alpha 1 (PDHA1), PDHB, and glutaminase (GLS) were predicted to target 111 miRNAs. PDHA1, FDX1, LIPT1, PDHB, LIAS, DLAT, GLS, and dihydrolipoamide dehydrogenase (DLD) were predicted to interact with 11, 10, 10, 9, 8, 7, 5, and 4 transcription factors, respectively. Finally, FDX1 expression was significantly upregulated in the hippocampus of ovariectomized rats with ischemia. Conclusion This study revealed an association between cerebral ischemic disease and cuproptosis, identifying eight potential target genes. These findings offer new insights into potential biomarkers for the diagnosis, treatment, and prognosis of cerebral ischemia, and provide avenues for the exploration of new medical intervention targets.
Collapse
Affiliation(s)
- Lihua Qin
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardiocerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xi Cao
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tengjia Huang
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yixin Liu
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Sheng Li
- Key Laboratory of Hunan Province for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardiocerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
6
|
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
|
7
|
Hao D, Luo W, Yan Y, Zhou J. Focus on cuproptosis: Exploring new mechanisms and therapeutic application prospects of cuproptosis regulation. Biomed Pharmacother 2024; 178:117182. [PMID: 39053428 DOI: 10.1016/j.biopha.2024.117182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Cuproptosis is a novel form of regulated cell death, which plays an important role in the physiological and pathological processes of the human body. Despite the increasing research on cuproptosis-related genes (CRGs) and their correlation with diseases, the pathogenesis of cuproptosis-related diseases remains unclear. Furthermore, there is a lack of reviews on the emerging technologies for regulating cuproptosis in disease treatment. This study delves into the copper-induced cell death mechanism, distinguishing cuproptosis from mechanisms like oxidative stress, glutathione synthesis inhibition, and ubiquitin-proteasome system inhibition. Several long-standing mysteries of diseases such as Wilson's disease and Menkes disease may be attributed to the occurrence of cuproptosis. In addition, we also review the detection indicators related to cuproptosis, providing targets for the diagnosis of cuproptosis-related diseases, and summarize the application value of cuproptosis in tumor therapy to better elucidate the impact of copper in cell death and diseases, and thus to promote the application prospects and possible strategies of cuproptosis-related substances, such as copper ion chelators, copper ion carriers, and copper nanomaterials, in disease therapy.
Collapse
Affiliation(s)
- Donglin Hao
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China; Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Wei Luo
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China; Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Yongmin Yan
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China; Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China.
| | - Jing Zhou
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China; Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China.
| |
Collapse
|
8
|
Lin CH, Chin Y, Zhou M, Sobol RW, Hung MC, Tan M. Protein lipoylation: mitochondria, cuproptosis, and beyond. Trends Biochem Sci 2024; 49:729-744. [PMID: 38714376 DOI: 10.1016/j.tibs.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 05/09/2024]
Abstract
Protein lipoylation, a crucial post-translational modification (PTM), plays a pivotal role in mitochondrial function and emerges as a key player in cell death through cuproptosis. This novel copper-driven cell death pathway is activated by excessive copper ions binding to lipoylated mitochondrial proteins, disrupting energy production and causing lethal protein aggregation and cell death. The intricate relationship among protein lipoylation, cellular energy metabolism, and cuproptosis offers a promising avenue for regulating essential cellular functions. This review focuses on the mechanisms of lipoylation and its significant impact on cell metabolism and cuproptosis, emphasizing the key genes involved and their implications for human diseases. It offers valuable insights into targeting dysregulated cellular metabolism for therapeutic purposes.
Collapse
Affiliation(s)
- Cheng-Han Lin
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan; Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical University, Taichung, Taiwan
| | - Yeh Chin
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan; Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical University, Taichung, Taiwan
| | - Ming Zhou
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Robert W Sobol
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School and Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
| | - Mien-Chie Hung
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan; Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical University, Taichung, Taiwan.
| | - Ming Tan
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan; Graduate Institute of Biomedical Sciences and Research Center for Cancer Biology, China Medical University, Taichung, Taiwan.
| |
Collapse
|
9
|
Gao X, Zhao H, Liu J, Wang M, Dai Z, Hao W, Wang Y, Wang X, Zhang M, Liu P, Cheng H, Liu Z. Enzalutamide Sensitizes Castration-Resistant Prostate Cancer to Copper-Mediated Cell Death. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401396. [PMID: 38859590 PMCID: PMC11321675 DOI: 10.1002/advs.202401396] [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: 02/07/2024] [Revised: 05/21/2024] [Indexed: 06/12/2024]
Abstract
Despite the initial efficacy of enzalutamide in castration-resistant prostate cancer (CRPC), inevitable resistance remains a significant challenge. Here, the synergistic induction of copper-dependent cell death (cuproptosis) in CRPC cells is reported by enzalutamide and copper ionophores (elesclomol/disulfiram). Mechanistically, enzalutamide treatment increases mitochondrial dependence in CRPC cells, rendering them susceptible to cuproptosis, as evidenced by specific reversal with the copper chelator tetrathiomolybdate. This susceptibility is characterized by hallmarks of cuproptosis, including lipoylated protein aggregation and iron-sulfur cluster protein instability. Interestingly, the mitochondrial matrix reductase, FDX1, specifically correlates with elesclomol sensitivity, suggesting a potential mechanistic divergence between the two copper ionophores. Notably, this synergistic effect extends beyond in vitro models, demonstrating efficacy in 22Rv1 xenografts, mouse Pten p53 knockout organoids. Importantly, enzalutamide significantly enhances copper ionophore-mediated cytotoxicity in enzalutamide-resistant cells. Collectively, these findings indicate that enzalutamide and copper ionophores synergistically induce cuproptosis, offering a promising therapeutic avenue for CRPC, potentially including enzalutamide-resistant cases.
Collapse
Affiliation(s)
- Xiang Gao
- Department of UrologySecond Hospital of Dalian Medical UniversityDalian116023China
| | - Haolin Zhao
- Department of UrologySecond Hospital of Dalian Medical UniversityDalian116023China
| | - Jiao Liu
- Dalian Key Laboratory of Molecular Targeted Cancer TherapyCancer InstituteThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Min Wang
- Dalian Key Laboratory of Molecular Targeted Cancer TherapyCancer InstituteThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Zhihong Dai
- Department of UrologySecond Hospital of Dalian Medical UniversityDalian116023China
| | - Wenjun Hao
- Department of UrologySecond Hospital of Dalian Medical UniversityDalian116023China
| | - Yanlong Wang
- Department of UrologySecond Hospital of Dalian Medical UniversityDalian116023China
| | - Xiang Wang
- Department of UrologySecond Hospital of Dalian Medical UniversityDalian116023China
| | - Min Zhang
- Dalian Key Laboratory of Molecular Targeted Cancer TherapyCancer InstituteThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Pixu Liu
- Dalian Key Laboratory of Molecular Targeted Cancer TherapyCancer InstituteThe Second Hospital of Dalian Medical UniversityDalian116023China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| | - Hailing Cheng
- Dalian Key Laboratory of Molecular Targeted Cancer TherapyCancer InstituteThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Zhiyu Liu
- Department of UrologySecond Hospital of Dalian Medical UniversityDalian116023China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological CancerDalian116023China
| |
Collapse
|
10
|
Xie XZ, Zuo L, Huang W, Fan QM, Weng YY, Yao WD, Jiang JL, Jin JQ. FDX1 as a novel biomarker and treatment target for stomach adenocarcinoma. World J Gastrointest Surg 2024; 16:1803-1824. [PMID: 38983344 PMCID: PMC11230022 DOI: 10.4240/wjgs.v16.i6.1803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is one of the main reasons for cancer-related deaths worldwide. This investigation aimed to define the connection between STAD and Cuproptosis-related genes (CRGs). Cuproptosis is a newly identified form of mitochondrial cell death triggered by copper. AIM To explore the identification of potential biomarkers for STAD disease based on cuproptosis. METHODS A predictive model using Gene Ontology (GO), Least Absolute Shrinkage and Selection Operator (LASSO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Variation Analysis (GSVA), and Gene Set Enrichment Analysis analyzed gene interconnections, focusing on 3 copper-related genes and their expression in The Cancer Genome Atlas-STAD. Networks for mRNA-miRNA and mRNA-transcription factor interactions were constructed. The prognostic significance of CRG scores was evaluated using time-receiver operating characteristic, Kaplan-Meier curves, and COX regression analysis. Validation was conducted with datasets GSE26942, GSE54129, and GSE66229. Expression of copper-related differentially expressed genes was also analyzed in various human tissues and gastric cancer subpopulations using the human protein atlas. RESULTS Three significant genes (FDX1, LIAS, MTF1) were identified and selected via LASSO analysis to predict and classify individuals with STAD into high and low CRG score subgroups. These genes were down-regulated in both risk categories. GO and KEGG analyses highlighted their involvement mainly in the electron transport chain. After validating their differential expression, FDX1 emerged as the most accurate diagnostic marker for gastric cancer. Additionally, the RCircos package localized FDX1 on chromosome 11. CONCLUSION Our study revealed that FDX1 could be a potential biomarker and treatment target for gastric malignancy, providing new ideas for further scientific research.
Collapse
Affiliation(s)
- Xian-Ze Xie
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Lei Zuo
- Anhui Province Huainan City Shou County Agricultural Machinery Affairs Management Center, Huainan 232200, Anhui Province, China
| | - Wei Huang
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Qiao-Mei Fan
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Ya-Yun Weng
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Wen-Dong Yao
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Jia-Li Jiang
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Jia-Qi Jin
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| |
Collapse
|
11
|
Jiang J, Hiron TK, Agbaedeng TA, Malhotra Y, Drydale E, Bancroft J, Ng E, Reschen ME, Davison LJ, O’Callaghan CA. A Novel Macrophage Subpopulation Conveys Increased Genetic Risk of Coronary Artery Disease. Circ Res 2024; 135:6-25. [PMID: 38747151 PMCID: PMC11191562 DOI: 10.1161/circresaha.123.324172] [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: 12/21/2023] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Coronary artery disease (CAD), the leading cause of death worldwide, is influenced by both environmental and genetic factors. Although over 250 genetic risk loci have been identified through genome-wide association studies, the specific causal variants and their regulatory mechanisms are still largely unknown, particularly in disease-relevant cell types such as macrophages. METHODS We utilized single-cell RNA-seq and single-cell multiomics approaches in primary human monocyte-derived macrophages to explore the transcriptional regulatory network involved in a critical pathogenic event of coronary atherosclerosis-the formation of lipid-laden foam cells. The relative genetic contribution to CAD was assessed by partitioning disease heritability across different macrophage subpopulations. Meta-analysis of single-cell RNA-seq data sets from 38 human atherosclerotic samples was conducted to provide high-resolution cross-referencing to macrophage subpopulations in vivo. RESULTS We identified 18 782 cis-regulatory elements by jointly profiling the gene expression and chromatin accessibility of >5000 macrophages. Integration with CAD genome-wide association study data prioritized 121 CAD-related genetic variants and 56 candidate causal genes. We showed that CAD heritability was not uniformly distributed and was particularly enriched in the gene programs of a novel CD52-hi lipid-handling macrophage subpopulation. These CD52-hi macrophages displayed significantly less lipoprotein accumulation and were also found in human atherosclerotic plaques. We investigated the cis-regulatory effect of a risk variant rs10488763 on FDX1, implicating the recruitment of AP-1 and C/EBP-β in the causal mechanisms at this locus. CONCLUSIONS Our results provide genetic evidence of the divergent roles of macrophage subsets in atherogenesis and highlight lipid-handling macrophages as a key subpopulation through which genetic variants operate to influence disease. These findings provide an unbiased framework for functional fine-mapping of genome-wide association study results using single-cell multiomics and offer new insights into the genotype-environment interactions underlying atherosclerotic disease.
Collapse
Affiliation(s)
- Jiahao Jiang
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Thomas K. Hiron
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Thomas A. Agbaedeng
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Yashaswat Malhotra
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Edward Drydale
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - James Bancroft
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Esther Ng
- Nuffield Department of Orthopaedics, Kennedy Institute of Rheumatology, Rheumatology and Musculoskeletal Sciences (E.N.), University of Oxford, United Kingdom
| | - Michael E. Reschen
- Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, United Kingdom (M.E.R.)
| | - Lucy J. Davison
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, United Kingdom (L.J.D.)
| | - Chris A. O’Callaghan
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| |
Collapse
|
12
|
Fang X, Ji Y, Li S, Wang L, He B, Li B, Liang B, Yin H, Chen H, Dingda D, Wu B, Gao F. Paeoniflorin attenuates cuproptosis and ameliorates left ventricular remodeling after AMI in hypobaric hypoxia environments. J Nat Med 2024; 78:664-676. [PMID: 38427210 PMCID: PMC11101588 DOI: 10.1007/s11418-024-01781-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/04/2024] [Indexed: 03/02/2024]
Abstract
This study investigates the cardioprotective effects of Paeoniflorin (PF) on left ventricular remodeling following acute myocardial infarction (AMI) under conditions of hypobaric hypoxia. Left ventricular remodeling post-AMI plays a pivotal role in exacerbating heart failure, especially at high altitudes. Using a rat model of AMI, the study aimed to evaluate the cardioprotective potential of PF under hypobaric hypoxia. Ninety male rats were divided into four groups: sham-operated controls under normoxia/hypobaria, an AMI model group, and a PF treatment group. PF was administered for 4 weeks after AMI induction. Left ventricular function was assessed using cardiac magnetic resonance imaging. Biochemical assays of cuproptosis, oxidative stress, apoptosis, inflammation, and fibrosis were performed. Results demonstrated PF significantly improved left ventricular function and remodeling after AMI under hypobaric hypoxia. Mechanistically, PF decreased FDX1/DLAT expression and serum copper while increasing pyruvate. It also attenuated apoptosis, inflammation, and fibrosis by modulating Bcl-2, Bax, NLRP3, and oxidative stress markers. Thus, PF exhibits therapeutic potential for left ventricular remodeling post-AMI at high altitude by inhibiting cuproptosis, inflammation, apoptosis and fibrosis. Further studies are warranted to optimize dosage and duration and elucidate PF's mechanisms of action.
Collapse
Affiliation(s)
- Xin Fang
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Road, Chengdu, 610041, China
| | - Yaoxuan Ji
- Department of Radiology, Ningxia Medical University, Yinchuan, China
| | - Shuang Li
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Road, Chengdu, 610041, China
| | - Lei Wang
- Molecular Imaging Center, West China Hospital, Sichuan University, Chengdu, China
| | - Bo He
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Road, Chengdu, 610041, China
| | - Bo Li
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Road, Chengdu, 610041, China
| | - Boshen Liang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Hongke Yin
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Road, Chengdu, 610041, China
| | - Haotian Chen
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Road, Chengdu, 610041, China
| | - Duojie Dingda
- Department of Radiology, Yushu People's Hospital, Yushu, Qinghai, China
| | - Bing Wu
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Road, Chengdu, 610041, China
| | - Fabao Gao
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Road, Chengdu, 610041, China.
| |
Collapse
|
13
|
Dieckmann CL. A hub for regulation of mitochondrial metabolism: Fatty acid and lipoic acid biosynthesis. IUBMB Life 2024; 76:332-344. [PMID: 38088214 DOI: 10.1002/iub.2802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/21/2023] [Indexed: 05/28/2024]
Abstract
Having evolved from a prokaryotic origin, mitochondria retain pathways required for the catabolism of energy-rich molecules and for the biosynthesis of molecules that aid catabolism and/or participate in other cellular processes essential for life of the cell. Reviewed here are details of the mitochondrial fatty acid biosynthetic pathway (FAS II) and its role in building both the octanoic acid precursor for lipoic acid biosynthesis (LAS) and longer-chain fatty acids functioning in chaperoning the assembly of mitochondrial multisubunit complexes. Also covered are the details of mitochondrial lipoic acid biosynthesis, which is distinct from that of prokaryotes, and the attachment of lipoic acid to subunits of pyruvate dehydrogenase, α-ketoglutarate dehydrogenase, and glycine cleavage system complexes. Special emphasis has been placed on presenting what is currently known about the interconnected paths and loops linking the FAS II-LAS pathway and two other mitochondrial realms, the organellar translation machinery and Fe-S cluster biosynthesis and function.
Collapse
Affiliation(s)
- Carol L Dieckmann
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
14
|
Liu M, Wu S, Wu H, Zhou Y, Zhang X, Zhu D, Jiang J. Ferredoxin 1: a gatekeeper in halting lung adenocarcinoma progression through activation of the GPRIN2 signaling pathway. J Transl Med 2024; 22:510. [PMID: 38802900 PMCID: PMC11131317 DOI: 10.1186/s12967-024-05277-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a highly lethal form of lung cancer. Despite advancements in treatments, managing LUAD is still challenging due to its aggressive behavior. Recent studies indicate that various molecular pathways, including the dysregulation of ferredoxin 1 (FDX1), play roles in LUAD progression. FDX1, a crucial protein in cellular redox reactions and energy metabolism, has been linked to several cancers. However, its exact role in the development of LUAD is not yet fully understood. METHODS We investigated the role of ferredoxin 1 (FDX1) in LUAD progression through analysis of its expression in LUAD tissues and its impact on patient survival. Functional assays were performed to assess the effects of FDX1 overexpression on LUAD cell proliferation, migration, and invasion. A xenograft model was employed to evaluate the tumorigenesis potential of LUAD cells with FDX1 overexpression. Mechanistic insights into FDX1 regulation were gained through depletion experiments targeting the G protein-regulated inducer of neurite outgrowth 2 (GPRIN2)/PI3K signaling pathway. RESULTS FDX1 expression was down-regulated in LUAD tissues, correlating with shorter patient survival. Overexpression of FDX1 suppressed LUAD cell proliferation, migration, and invasion in vitro, and inhibited tumorigenesis in vivo. Mechanistically, the GPRIN2/PI3K signaling pathway was implicated in FDX1 regulation, as depletion of GPRIN2 reversed the effects of FDX1 overexpression on cellular functions. CONCLUSIONS Our findings highlight FDX1 as a potential tumor suppressor in LUAD, acting through modulation of the GPRIN2/PI3K signaling pathway. These results suggest FDX1 as a promising therapeutic target for LUAD treatment, warranting further investigation into its clinical relevance.
Collapse
Affiliation(s)
- Ming Liu
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Shaoxian Wu
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Haoyu Wu
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - You Zhou
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Xinyu Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
- Dalian Medical University, Dalian, China
| | - Dawei Zhu
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| | - Jingting Jiang
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
15
|
Tang D, Kroemer G, Kang R. Targeting cuproplasia and cuproptosis in cancer. Nat Rev Clin Oncol 2024; 21:370-388. [PMID: 38486054 DOI: 10.1038/s41571-024-00876-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2024] [Indexed: 04/26/2024]
Abstract
Copper, an essential trace element that exists in oxidized and reduced forms, has pivotal roles in a variety of biological processes, including redox chemistry, enzymatic reactions, mitochondrial respiration, iron metabolism, autophagy and immune modulation; maintaining copper homeostasis is crucial as both its deficiency and its excess are deleterious. Dysregulated copper metabolism has a dual role in tumorigenesis and cancer therapy. Specifically, cuproplasia describes copper-dependent cell growth and proliferation, including hyperplasia, metaplasia and neoplasia, whereas cuproptosis refers to a mitochondrial pathway of cell death triggered by excessive copper exposure and subsequent proteotoxic stress (although complex interactions between cuproptosis and other cell death mechanisms, such as ferroptosis, are likely and remain enigmatic). In this Review, we summarize advances in our understanding of copper metabolism, the molecular machineries underlying cuproplasia and cuproptosis, and their potential targeting for cancer therapy. These new findings advance the rapidly expanding field of translational cancer research focused on metal compounds.
Collapse
Affiliation(s)
- Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM U1138, Equipe labellisée-Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
16
|
Jiayi H, Ziyuan T, Tianhua X, Mingyu Z, Yutong M, Jingyu W, Hongli Z, Li S. Copper homeostasis in chronic kidney disease and its crosstalk with ferroptosis. Pharmacol Res 2024; 202:107139. [PMID: 38484857 DOI: 10.1016/j.phrs.2024.107139] [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: 01/15/2024] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 03/27/2024]
Abstract
Chronic kidney disease (CKD) has become a global public health problem with high morbidity and mortality. Renal fibrosis can lead to end-stage renal disease (ESRD). However, there is still no effective treatment to prevent or delay the progression of CKD into ESRD. Therefore, exploring the pathogenesis of CKD is essential for preventing and treating CKD. There are a variety of trace elements in the human body that interact with each other within a complex regulatory network. Iron and copper are both vital trace elements in the body. They are critical for maintaining bodily functions, and the dysregulation of their metabolism can cause many diseases, including kidney disease. Ferroptosis is a new form of cell death characterized by iron accumulation and lipid peroxidation. Studies have shown that ferroptosis is closely related to kidney disease. However, the role of abnormal copper metabolism in kidney disease and its relationship with ferroptosis remains unclear. Here, our current knowledge regarding copper metabolism, its regulatory mechanism, and the role of abnormal copper metabolism in kidney diseases is summarized. In addition, we discuss the relationship between abnormal copper metabolism and ferroptosis to explore the possible pathogenesis and provide a potential therapeutic target for CKD.
Collapse
Affiliation(s)
- Huang Jiayi
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Tong Ziyuan
- China Medical University, Shenyang 110122, People's Republic of China
| | - Xu Tianhua
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Zhang Mingyu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Ma Yutong
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Wang Jingyu
- Renal Division, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Zhou Hongli
- Department of Nephrology, The First Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province 110004, People's Republic of China
| | - Sun Li
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.
| |
Collapse
|
17
|
Moison C, Gracias D, Schmitt J, Girard S, Spinella JF, Fortier S, Boivin I, Mendoza-Sanchez R, Thavonekham B, MacRae T, Mayotte N, Bonneil E, Wittman M, Carmichael J, Ruel R, Thibault P, Hébert J, Marinier A, Sauvageau G. SF3B1 mutations provide genetic vulnerability to copper ionophores in human acute myeloid leukemia. SCIENCE ADVANCES 2024; 10:eadl4018. [PMID: 38517966 PMCID: PMC10959413 DOI: 10.1126/sciadv.adl4018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/20/2024] [Indexed: 03/24/2024]
Abstract
In a phenotypical screen of 56 acute myeloid leukemia (AML) patient samples and using a library of 10,000 compounds, we identified a hit with increased sensitivity toward SF3B1-mutated and adverse risk AMLs. Through structure-activity relationship studies, this hit was optimized into a potent, specific, and nongenotoxic molecule called UM4118. We demonstrated that UM4118 acts as a copper ionophore that initiates a mitochondrial-based noncanonical form of cell death known as cuproptosis. CRISPR-Cas9 loss-of-function screen further revealed that iron-sulfur cluster (ISC) deficiency enhances copper-mediated cell death. Specifically, we found that loss of the mitochondrial ISC transporter ABCB7 is synthetic lethal to UM4118. ABCB7 is misspliced and down-regulated in SF3B1-mutated leukemia, creating a vulnerability to copper ionophores. Accordingly, ABCB7 overexpression partially rescued SF3B1-mutated cells to copper overload. Together, our work provides mechanistic insights that link ISC deficiency to cuproptosis, as exemplified by the high sensitivity of SF3B1-mutated AMLs. We thus propose SF3B1 mutations as a biomarker for future copper ionophore-based therapies.
Collapse
Affiliation(s)
- Céline Moison
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Deanne Gracias
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Julie Schmitt
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Simon Girard
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Jean-François Spinella
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Simon Fortier
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Isabel Boivin
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | | | - Bounkham Thavonekham
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Tara MacRae
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Nadine Mayotte
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Eric Bonneil
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Mark Wittman
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA, USA
| | - James Carmichael
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA, USA
| | - Réjean Ruel
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Pierre Thibault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Department of Chemistry, Université de Montréal, Montréal, Canada
| | - Josée Hébert
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Division of Hematology-Oncology and Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Anne Marinier
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Department of Chemistry, Université de Montréal, Montréal, Canada
| | - Guy Sauvageau
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Division of Hematology-Oncology and Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| |
Collapse
|
18
|
Gonzalez L, Chau-Duy Tam Vo S, Faivre B, Pierrel F, Fontecave M, Hamdane D, Lombard M. Activation of Coq6p, a FAD Monooxygenase Involved in Coenzyme Q Biosynthesis, by Adrenodoxin Reductase/Ferredoxin. Chembiochem 2024; 25:e202300738. [PMID: 38141230 DOI: 10.1002/cbic.202300738] [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: 10/26/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 12/25/2023]
Abstract
Adrenodoxin reductase (AdxR) plays a pivotal role in electron transfer, shuttling electrons between NADPH and iron/sulfur adrenodoxin proteins in mitochondria. This electron transport system is essential for P450 enzymes involved in various endogenous biomolecules biosynthesis. Here, we present an in-depth examination of the kinetics governing the reduction of human AdxR by NADH or NADPH. Our results highlight the efficiency of human AdxR when utilizing NADPH as a flavin reducing agent. Nevertheless, akin to related flavoenzymes such as cytochrome P450 reductase, we observe that low NADPH concentrations hinder flavin reduction due to intricate equilibrium reactions between the enzyme and its substrate/product. Remarkably, the presence of MgCl2 suppresses this complex kinetic behavior by decreasing NADPH binding to oxidized AdxR, effectively transforming AdxR into a classical Michaelis-Menten enzyme. We propose that the addition of MgCl2 may be adapted for studying the reductive half-reactions of other flavoenzymes with NADPH. Furthermore, in vitro experiments provide evidence that the reduction of the yeast flavin monooxygenase Coq6p relies on an electron transfer chain comprising NADPH-AdxR-Yah1p-Coq6p, where Yah1p shuttles electrons between AdxR and Coq6p. This discovery explains the previous in vivo observation that Yah1p and the AdxR homolog, Arh1p, are required for the biosynthesis of coenzyme Q in yeast.
Collapse
Affiliation(s)
- Lucie Gonzalez
- Laboratoire de Chimie des Processus Biologiques, Collège de France, Sorbonne Université, CNRS UMR8229, PSL Research University, Sorbonne Université, 11 place Marcelin Berthelot, 75 005, Paris, France
| | - Samuel Chau-Duy Tam Vo
- Laboratoire de Chimie des Processus Biologiques, Collège de France, Sorbonne Université, CNRS UMR8229, PSL Research University, Sorbonne Université, 11 place Marcelin Berthelot, 75 005, Paris, France
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bruno Faivre
- Laboratoire de Chimie des Processus Biologiques, Collège de France, Sorbonne Université, CNRS UMR8229, PSL Research University, Sorbonne Université, 11 place Marcelin Berthelot, 75 005, Paris, France
| | - Fabien Pierrel
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, 38000, Grenoble, France
| | - Marc Fontecave
- Laboratoire de Chimie des Processus Biologiques, Collège de France, Sorbonne Université, CNRS UMR8229, PSL Research University, Sorbonne Université, 11 place Marcelin Berthelot, 75 005, Paris, France
| | - Djemel Hamdane
- Laboratoire de Chimie des Processus Biologiques, Collège de France, Sorbonne Université, CNRS UMR8229, PSL Research University, Sorbonne Université, 11 place Marcelin Berthelot, 75 005, Paris, France
- Institut de Biologie Paris-Seine, Biology of Aging and Adaptation, UMR 8256, Sorbonne Université, 7 quai Saint-Bernard, 75 252, Paris, France
| | - Murielle Lombard
- Laboratoire de Chimie des Processus Biologiques, Collège de France, Sorbonne Université, CNRS UMR8229, PSL Research University, Sorbonne Université, 11 place Marcelin Berthelot, 75 005, Paris, France
| |
Collapse
|
19
|
Zhong Y, Zeng W, Chen Y, Zhu X. The effect of lipid metabolism on cuproptosis-inducing cancer therapy. Biomed Pharmacother 2024; 172:116247. [PMID: 38330710 DOI: 10.1016/j.biopha.2024.116247] [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: 12/09/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Cuproptosis provides a new therapeutic strategy for cancer treatment and is thought to have broad clinical application prospects. Nevertheless, some oncological clinical trials have yet to demonstrate favorable outcomes, highlighting the need for further research into the molecular mechanisms underlying cuproptosis in tumors. Cuproptosis primarily hinges on the intracellular accumulation of copper, with lipid metabolism exerting a profound influence on its course. The interaction between copper metabolism and lipid metabolism is closely related to cuproptosis. Copper imbalance can affect mitochondrial respiration and lipid metabolism changes, while lipid accumulation can promote copper uptake and absorption, and inhibit cuproptosis induced by copper. Anomalies in lipid metabolism can disrupt copper homeostasis within cells, potentially triggering cuproptosis. The interaction between cuproptosis and lipid metabolism regulates the occurrence, development, metastasis, chemotherapy drug resistance, and tumor immunity of cancer. Cuproptosis is a promising new target for cancer treatment. However, the influence of lipid metabolism and other factors should be taken into consideration. This review provides a brief overview of the characteristics of the interaction between cuproptosis and lipid metabolism in cancer and analyses potential strategies of applying cuproptosis for cancer treatment.
Collapse
Affiliation(s)
- Yue Zhong
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Wei Zeng
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yongbo Chen
- Rehabilitation College of Gannan Medical University, Ganzhou 341000, China
| | - Xiuzhi Zhu
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
20
|
魏 婷, 丁 洋, 张 佳, 李 金, 张 恒, 康 品, 张 宁. [Correlation of serum ferredoxin 1 and lipoic acid levels with severity of coronary artery disease]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:308-316. [PMID: 38501416 PMCID: PMC10954524 DOI: 10.12122/j.issn.1673-4254.2024.02.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Indexed: 03/20/2024]
Abstract
OBJECTIVE To analyze the correlation of copper death inducer ferredoxin 1 (FDX1) and lipoic acid (LA) with the occurrence and severity of coronary atherosclerosis and explore their roles in coronary heart disease (CHD). METHODS We analyzed the data of 226 patients undergoing coronary artery angiography (CAG) in our hospital between October, 2021 and October, 2022, including 47 patients with normal CAG findings (control group) and 179 patients with mild, moderate or severe coronary artery stenosis (CHD group). Serum FDX1 and LA levels were determined with ELISA for all the patients. We also examined pathological changes in the aorta of normal and ApoE-/- mice using HE staining and observed collagen fiber deposition with Sirius red staining. Immunohistochemistry was used to detect the expression and distribution of FDX1 and LA in the aorta, and RT-PCR was performed to detect the expressions of FDX1, LIAS and ACO2 mRNAs in the myocardial tissues. RESULTS Compared with the control patients, CHD patients had significantly lower serum FDX1 and LA levels, which decreased progressively as coronary artery stenosis worsened (P < 0.01) and as the number of involved coronary artery branches increased (P < 0.05). Serum FDX1 and LA levels were positively correlated (r=0.451, P < 0.01) and they both negatively correlated with the Gensini score (r=-0.241 and -0.273, respectively; P < 0.01). Compared with normal mice, ApoE-/- mice showed significantly increased lipid levels (P < 0.01) and atherosclerosis index, obvious thickening, lipid aggregation, and collagen fiber hyperplasia in the aorta, and significantly reduced expressions of FDX1, LA, LIAS, and ACO2 (P < 0.05). CONCLUSION Serum FDX1 and LA levels decrease with worsening of coronary artery lesions, and theirs expressions are correlated with coronary artery lesions induced by hyperlipidemia.
Collapse
Affiliation(s)
- 婷 魏
- 蚌埠医科大学第一附属医院心血管科,安徽 蚌埠 233000Department of Cardiovascular Medicine of First Affiliated Hospital, Bengbu Medical University, Bengbu 233000, China
| | - 洋洋 丁
- 蚌埠医科大学第一附属医院心血管科,安徽 蚌埠 233000Department of Cardiovascular Medicine of First Affiliated Hospital, Bengbu Medical University, Bengbu 233000, China
| | - 佳佳 张
- 蚌埠医科大学第一附属医院心血管科,安徽 蚌埠 233000Department of Cardiovascular Medicine of First Affiliated Hospital, Bengbu Medical University, Bengbu 233000, China
| | - 金龙 李
- 蚌埠医科大学第一附属医院心血管科,安徽 蚌埠 233000Department of Cardiovascular Medicine of First Affiliated Hospital, Bengbu Medical University, Bengbu 233000, China
| | - 恒 张
- 蚌埠医科大学第一附属医院心血管科,安徽 蚌埠 233000Department of Cardiovascular Medicine of First Affiliated Hospital, Bengbu Medical University, Bengbu 233000, China
| | - 品方 康
- 蚌埠医科大学第一附属医院心血管科,安徽 蚌埠 233000Department of Cardiovascular Medicine of First Affiliated Hospital, Bengbu Medical University, Bengbu 233000, China
- 蚌埠医科大学心脑血管基础与临床重点实验室,安徽 蚌埠 233000Key Laboratory of Preclinical and Clinical Cardiovascular Medicine, Bengbu Medical University, Bengbu 233000, China
| | - 宁汝 张
- 蚌埠医科大学第一附属医院心血管科,安徽 蚌埠 233000Department of Cardiovascular Medicine of First Affiliated Hospital, Bengbu Medical University, Bengbu 233000, China
| |
Collapse
|
21
|
Yang M, Wang Y, He L, Shi X, Huang S. Comprehensive bioinformatics analysis reveals the role of cuproptosis-related gene Ube2d3 in myocardial infarction. Front Immunol 2024; 15:1353111. [PMID: 38440726 PMCID: PMC10909922 DOI: 10.3389/fimmu.2024.1353111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Background Myocardial infarction (MI) caused by severe coronary artery disease has high incidence and mortality rates, making its prevention and treatment a central and challenging aspect of clinical work for cardiovascular practitioners. Recently, researchers have turned their attention to a novel mechanism of cell death caused by Cu2+, cuproptosis. Methods This study integrated data from three MI-related bulk datasets downloaded from the Gene Expression Omnibus (GEO) database, and identified 16 differentially expressed genes (DEGs) related to cuproptosis by taking intersection of the 6378 DEGs obtained by differential analysis with 49 cuproptosis-related genes. Four hub genes, Dbt, Dlat, Ube2d1 and Ube2d3, were screened out through random forest analysis and Lasso analysis. In the disease group, Dbt, Dlat, and Ube2d1 showed low expression, while Ube2d3 exhibited high expression. Results Focusing on Ube2d3 for subsequent functional studies, we confirmed its high expression in the MI group through qRT-PCR and Western Blot detection after successful construction of a MI mouse model by left anterior descending (LAD) coronary artery ligation, and further clarified the correlation of cuproptosis with MI development by detecting the levels of cuproptosis-related proteins. Moreover, through in vitro experiments, Ube2d3 was confirmed to be highly expressed in oxygen-glucose deprivation (OGD)-treated cardiomyocytes AC16. In order to further clarify the role of Ube2d3, we knocked down Ube2d3 expression in OGD-treated AC16 cells, and confirmed Ube2d3's promoting role in the hypoxia damage of AC16 cells by inducing cuproptosis, as evidenced by the detection of MTT, TUNEL, LDH release and cuproptosis-related proteins. Conclusion In summary, our findings indicate that Ube2d3 regulates cuproptosis to affect the progression of MI.
Collapse
Affiliation(s)
- Ming Yang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yucheng Wang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liming He
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinxin Shi
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuwei Huang
- Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, China
| |
Collapse
|
22
|
Liu N, Chen M. Crosstalk between ferroptosis and cuproptosis: From mechanism to potential clinical application. Biomed Pharmacother 2024; 171:116115. [PMID: 38181713 DOI: 10.1016/j.biopha.2023.116115] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/23/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
Ferroptosis and cuproptosis, regulated forms of cell death resulting from metal ion accumulation, are closely related in terms of occurrence, cell metabolism, signaling pathways, and drug resistance. Notably, it is now understood that these processes play crucial roles in regulating physiological and pathological processes, especially in tumor development. Consequently, ferroptosis and cuproptosis have gained increasing significance as potential targets for anti-cancer drug development. This article systematically outlines the molecular mechanisms and cross-talk components of both ferroptosis and cuproptosis, elucidating their impacts on cancer. Furthermore, it investigates the clinical perspective of targeted ferroptosis and cuproptosis in cancer chemotherapy, immunotherapy, and radiotherapy. Our discussion extends to a comparative analysis of nanoparticles developed based on the mechanisms of ferroptosis and cuproptosis in cancer, contrasting them with current conventional therapies. Opportunities and challenges in cancer treatment are explored, emphasizing the potential therapeutic direction of co-targeting ferroptosis and cuproptosis. The article also attempts to analyze the clinical applications of this co-targeting approach for cancer treatment while summarizing the existing barriers that require overcoming.
Collapse
Affiliation(s)
- Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Minbin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
23
|
Liu WQ, Lin WR, Yan L, Xu WH, Yang J. Copper homeostasis and cuproptosis in cancer immunity and therapy. Immunol Rev 2024; 321:211-227. [PMID: 37715546 DOI: 10.1111/imr.13276] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/17/2023]
Abstract
Copper is an essential nutrient for maintaining enzyme activity and transcription factor function. Excess copper results in the aggregation of lipoylated dihydrolipoamide S-acetyltransferase (DLAT), which correlates to the mitochondrial tricarboxylic acid (TCA) cycle, resulting in proteotoxic stress and eliciting a novel cell death modality: cuproptosis. Cuproptosis exerts an indispensable role in cancer progression, which is considered a promising strategy for cancer therapy. Cancer immunotherapy has gained extensive attention owing to breakthroughs in immune checkpoint blockade; furthermore, cuproptosis is strongly connected to the modulation of antitumor immunity. Thus, a thorough recognition concerning the mechanisms involved in the modulation of copper metabolism and cuproptosis may facilitate improvement in cancer management. This review outlines the cellular and molecular mechanisms and characteristics of cuproptosis and the links of the novel regulated cell death modality with human cancers. We also review the current knowledge on the complex effects of cuproptosis on antitumor immunity and immune response. Furthermore, potential agents that elicit cuproptosis pathways are summarized. Lastly, we discuss the influence of cuproptosis induction on the tumor microenvironment as well as the challenges of adding cuproptosis regulators to therapeutic strategies beyond traditional therapy.
Collapse
Affiliation(s)
- Wei-Qing Liu
- Department of Internal Medicine-Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wan-Rong Lin
- Department of Surgical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Yan
- Department of Internal Medicine-Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wen-Hao Xu
- Department of Internal Medicine-Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Yang
- Department of Surgical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
24
|
Qian S, Liu J, Liao W, Wang F. METTL3 promotes non-small-cell lung cancer growth and metastasis by inhibiting FDX1 through copper death-associated pri-miR-21-5p maturation. Epigenomics 2023; 15:1237-1255. [PMID: 38126112 DOI: 10.2217/epi-2023-0230] [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] [Indexed: 12/23/2023] Open
Abstract
Objective: We probed into the significance of METTL3 in the maturation process of pri-miR-21-5p. We specifically investigated its impact on the regulation of FDX1 and its involvement in the progression of non-small-cell lung cancer (NSCLC). Methods: The Cancer Genome Atlas (TCGA) identified NSCLC factors. Methylation-specific PCR (MSP), clonogenic tests and flow cytometry analyzed cells. Methylated RNA immunoprecipitation (Me-RIP) and dual-luciferase studied miR-21-5p/FDX1. Mice xenografts showed METTL3's tumorigenic effect. Results: METTL3, with high expression but low methylation in NSCLC, influenced cell behaviors. Its suppression reduced oncogenic properties. METTL3 enhanced miR-21-5p maturation, targeting FDX1 and boosting NSCLC tumorigenicity in mice. Conclusion: METTL3 may promote NSCLC development by facilitating pri-miR-21-5p maturation, upregulating miR-21-5p and targeting inhibition of FDX1.
Collapse
Affiliation(s)
- Shuai Qian
- Department of Clinical Laboratory, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, People's Republic of China
| | - Jun Liu
- Department of Clinical Laboratory, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, People's Republic of China
| | - Wenliang Liao
- Department of Clinical Laboratory, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, People's Republic of China
| | - Fengping Wang
- Department of Clinical Laboratory, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, People's Republic of China
| |
Collapse
|
25
|
Liu X, Luo B, Wu X, Tang Z. Cuproptosis and cuproptosis-related genes: Emerging potential therapeutic targets in breast cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:189013. [PMID: 37918452 DOI: 10.1016/j.bbcan.2023.189013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/12/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
Breast cancer is one of the most common malignant tumors in women worldwide, and thus, it is important to enhance its treatment efficacy [1]. Copper has emerged as a critical trace element that affects various intracellular signaling pathways, gene expression, and biological metabolic processes [2], thereby playing a crucial role in the pathogenesis of breast cancer. Recent studies have identified cuproptosis, a newly discovered type of cell death, as an emerging therapeutic target for breast cancer treatment, thereby offering new hope for breast cancer patients. Tsvetkov's research has elucidated the mechanism of cuproptosis and uncovered the critical genes involved in its regulation [3]. Manipulating the expression of these genes could potentially serve as a promising therapeutic strategy for breast cancer treatment. Additionally, using copper ionophores and copper complexes combined with nanomaterials to induce cuproptosis may provide a potential approach to eliminating drug-resistant breast cancer cells, thus improving the therapeutic efficacy of chemotherapy, radiotherapy, and immunotherapy and eventually eradicating breast tumors. This review aims to highlight the practical significance of cuproptosis-related genes and the induction of cuproptosis in the clinical diagnosis and treatment of breast cancer. We examine the potential of cuproptosis as a novel therapeutic target for breast cancer, and we explore the present challenges and limitations of this approach. Our objective is to provide innovative ideas and references for the development of breast cancer treatment strategies based on cuproptosis.
Collapse
Affiliation(s)
- Xiangdong Liu
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, Wuhan 430079, China
| | - Bo Luo
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, Wuhan 430079, China.
| | - Xinhong Wu
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, Wuhan 430079, China
| | - Zijian Tang
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|