1
|
Zhang W, Lin YT, Jiang JW, Hu Y. Mapping the expression and functional landscape of key enzymes in glucose metabolism within human gynecological tumors. Curr Probl Cancer 2025; 54:101155. [PMID: 39521683 DOI: 10.1016/j.currproblcancer.2024.101155] [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: 07/26/2024] [Revised: 10/14/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Gynecological tumors, primarily ovarian cancer (OC), cervical cancer (CC), and endometrial cancer (EC), have a significant global impact on women's health, characterized by high mortality rates. Emerging evidence underscores the pivotal role of altered glucose metabolism in the initiation and progression of these malignancies. Glucose metabolism, encompassing glycolysis, the tricarboxylic acid (TCA) cycle, oxidative phosphorylation, and the pentose phosphate pathway (PPP), among others, is intricately governed by a spectrum of key enzymes. These enzymes drive metabolic reprogramming essential for tumor growth and survival, thereby influencing patient outcomes and clinical management strategies. However, the comprehensive characterization and summary of the expression profiles, regulatory networks involved, and functional roles of these glucose metabolic enzymes in human gynecological tumors remain incomplete. In this review, we systematically map the expression landscape of these critical glucose metabolic enzymes in gynecological cancers based on research utilizing clinical gynecological tumor tissues. Additionally, we summarize the specific functions of key enzymes of glucose metabolism and the pathways they regulate in gynecological tumors. This review provides profound insights into the metabolic dynamics underlying these diseases. This understanding illuminates the metabolic strategies employed by tumor cells and sets the stage for innovative therapeutic approaches targeting cancer cell glucose metabolic dependencies, thereby holding promise for enhancing patient outcomes in gynecological oncology.
Collapse
Affiliation(s)
- Wen Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China; Department of Ultrasonic Medicine, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Ying-Tong Lin
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jing-Wen Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yan Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Cong Y, Li N, Zhang Z, Shang Y, Zhao H. Cuproptosis: molecular mechanisms, cancer prognosis, and therapeutic applications. J Transl Med 2025; 23:104. [PMID: 39844182 PMCID: PMC11752808 DOI: 10.1186/s12967-025-06121-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025] Open
Abstract
Cuproptosis differs from other forms of cell death, such as apoptosis, necroptosis, and ferroptosis, in its unique molecular mechanisms and signaling pathways. In this review, we delve into the cellular metabolic pathways of copper, highlighting the role of copper in biomolecule synthesis, mitochondrial respiration, and antioxidant defense. Furthermore, we elucidate the relationship between cuproptosis-related genes (CRGs) and cancer prognosis, analyzing their expression patterns across various tumor types and their impact on patient outcomes. Our review also uncovers the potential therapeutic applications of copper chelators, copper ionophores, and copper-based nanomaterials in oncology. In addition, we discuss the emerging role of cuproptosis in remodeling the tumor microenvironment, enhancing immune cell infiltration, and converting "cold tumors" into "hot tumors" that respond better to immunotherapy. In short, this review underscores the pivotal importance of cuproptosis in cancer biology and highlights its translational potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Yating Cong
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Na Li
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Zixin Zhang
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Yan Shang
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Hailong Zhao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| |
Collapse
|
3
|
Cui Y, Luo S, Wu B, Li Q, Han F, Wang Z. Immunomodulatory Effects of SPHK1 and Its Interaction with TFAP2A in Yellow Drum ( Nibea albiflora). Int J Mol Sci 2024; 25:13641. [PMID: 39769404 PMCID: PMC11728317 DOI: 10.3390/ijms252413641] [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: 11/05/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Sphingosine kinases (SPHKs) are essential enzymes that catalyze the phosphorylation of sphingosine to produce sphingosine-1-phosphate (S1P), which plays pivotal roles in inflammation and immune regulation. In this study, genome-wide association analysis (GWAS) identified the Ydsphk1 gene as closely associated with the resistance of yellow drum (Nibea albiflora) to Vibrio harveyi. Structural prediction showed that YDSPHK1 contains a typical diacylglycerol kinase catalytic (DAGKc) domain (154-291 aa). By constructing and transfecting Ydsphk1 expression plasmids into yellow drum kidney cells, we found that YDSPHK1 is localized in the cytoplasm. Subsequent RNA-Seq analysis of an overexpression plasmid identified 25 differentially expressed genes (DEGs), including 13 upregulated and 12 downregulated. Notably, nsun5 and hsp90aa1 were significantly upregulated, while Nfkbia and hmox1 were downregulated. Promoter analysis indicated that the core regulatory regions of Ydsphk1 are located between -1931~-1679 bp and -419~+92 bp, with two predicted TFAP2A binding sites in the -419~+92 bp region. Further studies demonstrated that varying concentrations of TFAP2A significantly reduced Ydsphk1 promoter activity. These findings underscore the pivotal role of Ydsphk1 in regulating immune responses in yellow drum, particularly through its impact on key immune-related genes and pathways such as NF-κB signaling and ferroptosis. The identification of Ydsphk1 as a mediator of immune regulation provides valuable insights into the molecular mechanisms of immune defense and highlights its potential as a target for enhancing pathogen resistance in aquaculture practices. This study lays a strong foundation for future research aimed at developing innovative strategies for disease management in aquaculture species.
Collapse
Affiliation(s)
- Yu Cui
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Shuai Luo
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Baolan Wu
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Qiaoying Li
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Fang Han
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Zhiyong Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
4
|
Li Z, Li N, Ndzie Noah ML, Shao Q, Zhan X. Pharmacoproteomics reveals energy metabolism pathways as therapeutic targets of ivermectin in ovarian cancer toward 3P medical approaches. EPMA J 2024; 15:711-737. [PMID: 39635022 PMCID: PMC11612093 DOI: 10.1007/s13167-024-00385-1] [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: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Objective Ovarian cancer is the malignant tumor with the highest mortality rate in the female reproductive system, enormous socio-economic burden, and limited effective drug therapy. There is an urgent need to find novel effective drugs for ovarian cancer therapy. Our previous in vitro studies demonstrate that ivermectin effectively inhibits ovarian cancer cells and affects energy metabolism pathways. This study aims to clarify in vivo mechanisms and therapeutic targets of ivermectin in the treatment of ovarian cancer to establish predictive biomarkers, guide personalized treatments, and improve preventive strategies in the framework of 3P medicine. Methods A TOV-21G tumor-bearing mouse model was constructed based on histopathological data and biochemical parameters. TMT-based proteomic analysis was performed on tumor tissues from the different treatment groups. All significantly differentially abundant proteins were characterized by hierarchical clustering, Gene Ontology (GO) enrichment analyses, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. In addition, the data were integrated and analyzed with the proteomic data of clinical ovarian cancer tissues from our previous study and the proteomic data of ivermectin intervention in ovarian cancer cells to identify key regulators of ivermectin. Results Ivermectin (10 mg/kg) had a significant anti-ovarian cancer effect in mice, with a tumor inhibitory rate of 61.5%. Molecular changes in tumor tissue of ivermectin-treated mice were established, and protein-protein interaction (PPI) analysis showed that the main differential pathway networks included the TCA cycle, propanoate metabolism, 2-0xocarboxyacid metabolism, and other pathways. Integrating our previous clinical ovarian cancer tissue and cell experimental data, this study found that ivermectin significantly interfered with the energy metabolic pathways of ovarian cancer, including glycolysis, TCA cycle, oxidative phosphorylation, and other related pathways. Conclusions This study evaluated the anti-ovarian cancer effect in vitro and in vivo, and its specific regulatory effect on energy metabolism. The expressions of drug target molecules in the energy metabolism pathway of ovarian cancer will be used to guide the diagnosis and prevention of ovarian cancer. The significant efficacy of ivermectin will be applied to the treatment of ovarian cancer and personalized medication. This has guiding significance for the clinical diagnosis, treatment, personalized medication, and prognosis evaluation of ovarian cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00385-1.
Collapse
Affiliation(s)
- Zhijun Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Marie Louise Ndzie Noah
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Qianwen Shao
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
5
|
Zhang X, Shi X, Zhang X, Zhang Y, Yu S, Zhang Y, Liu Y. Repositioning fluphenazine as a cuproptosis-dependent anti-breast cancer drug candidate based on TCGA database. Biomed Pharmacother 2024; 178:117293. [PMID: 39142251 DOI: 10.1016/j.biopha.2024.117293] [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: 06/01/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024] Open
Abstract
Breast cancer is one of the most prevalent malignancies among women. Enhancing the prognosis is an effective approach to enhance the survival rate of breast cancer. Cuproptosis, a copper-dependent programmed cell death process, has been associated with patient prognosis. Inducing cuproptosis is a promising approach for therapy. However, there is currently no anti-breast cancer drug that induces cuproptosis. In this study, we repositioned the clinical drug fluphenazine as a potential agent for breast cancer treatment by inducing cuproptosis. Firstly, we utilized the Cancer Genome Atlas (TCGA) database and Connectivity Map (CMap) database to identify 22 potential compounds with anti-breast cancer activity through inducing cuproptosis. Subsequently, our findings demonstrated that fluphenazine effectively suppressed the viability of MCF-7 cells. Fluphenazine also significantly inhibited the viability of triple negative breast cancer cells MDA-MB-453 and MDA-MB-231. Furthermore, our study revealed that fluphenazine significantly down-regulated the expression of potential prognostic biomarkers associated with cuproptosis, increased copper ion levels, and reduced intracellular pyruvate accumulation. Additionally, it up-regulated the expression of FDX1 at both the mRNA and protein levels, which has been reported to play a crucial role in the induction of cuproptosis. These findings suggest that fluphenazine has the potential to be used as an anti-breast cancer drug by inducing cuproptosis. Therefore, this research provides an insight for the development of novel cuproptosis-dependent anti-cancer agents.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Endocrinology, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China; Department of Pharmacology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaoyuan Shi
- Department of Pharmacology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Xi Zhang
- Department of Pharmacology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Ying Zhang
- Department of Pharmacology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Siting Yu
- Department of Pharmacology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Yi Zhang
- Department of Endocrinology, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China; Department of Pharmacology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030001, China; School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China.
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
6
|
Shi M, Mobet Y, Shen H. Quercetin Attenuates Acute Kidney Injury Caused by Cisplatin by Inhibiting Ferroptosis and Cuproptosis. Cell Biochem Biophys 2024; 82:2687-2699. [PMID: 39026057 DOI: 10.1007/s12013-024-01379-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2024] [Indexed: 07/20/2024]
Abstract
Ferroptosis, an iron- and ROS-dependent form of regulated cell death. Cuproptosis is a novel form of cellular demise mode. Quercetin, a natural flavonoid, has demonstrated a range of pharmacological activities, including anti-cancer, anti-inflammatory, and antioxidant properties. In this research, we investigated the quercetin effect on cisplatin-induced acute kidney and its mechanism associated ferroptosis and cuproptosis. The HK-2 cells were used in this research. Cell viability was evaluated using the CCK-8 assay. Acute kidney injury (AKI) models were established to perform in vivo experiments. Renal tissue homogenate was used to determine ROS, LPO, MDA, PA, etc., to assess ferroptosis and cuproptosis. To perform bioinformatic analysis, microarray data from the GEO database was utilized. Real-time PCR analysis and ELISA was explored the mechanism of ferroptosis and cuproptosis. We found that ferroptosis and cuproptosis in AKI were abnormally activated caused by cisplatin, and that quercetin attenuated AKI by inhibiting ferroptosis and cuproptosis. QCT suppressed ferroptosis by reducing malondialdehyde (MDA) and ROS levels and increasing glutathione (GSH) levels and alleviated cuproptosis by reducing copper ion, pyruvate (PA) and HSP70 levels. Moreover, bioinformatic analysis revealed that the ferroptosis-related gene SLC7A11 and the cuproptosis-related genes ATP7B and GLS were the differential expression genes. And QCT significantly increased the expression or activity of SLC7A11, GPX4, ATP7B, and GLS in Cis-AKI mice. Our findings highlight the clinical importance of quercetin, which guards against cisplatin-induced acute kidney injury by suppressing ferroptosis and cuproptosis.
Collapse
Affiliation(s)
- Mengqi Shi
- Medical College, South China University of Technology, Guangzhou, 510006, China
| | - Youchaou Mobet
- Bai sheng Biological products Co. Ltd, Guangzhou, 511462, China
- NanSha Post Doctoral Research Workstation, Guangzhou, 511400, China
| | - Hong Shen
- Medical College, South China University of Technology, Guangzhou, 510006, China.
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
7
|
Nisco A, Tolomeo M, Scalise M, Zanier K, Barile M. Exploring the impact of flavin homeostasis on cancer cell metabolism. Biochim Biophys Acta Rev Cancer 2024; 1879:189149. [PMID: 38971209 DOI: 10.1016/j.bbcan.2024.189149] [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: 04/24/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Flavins and their associated proteins have recently emerged as compelling players in the landscape of cancer biology. Flavins, encompassing flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), serve as coenzymes in a multitude of cellular processes, such as metabolism, apoptosis, and cell proliferation. Their involvement in oxidative phosphorylation, redox homeostasis, and enzymatic reactions has long been recognized. However, recent research has unveiled an extended role for flavins in the context of cancer. In parallel, riboflavin transporters (RFVTs), FAD synthase (FADS), and riboflavin kinase (RFK) have gained prominence in cancer research. These proteins, responsible for riboflavin uptake, FAD biosynthesis, and FMN generation, are integral components of the cellular machinery that governs flavin homeostasis. Dysregulation in the expression/function of these proteins has been associated with various cancers, underscoring their potential as diagnostic markers, therapeutic targets, and key determinants of cancer cell behavior. This review embarks on a comprehensive exploration of the multifaceted role of flavins and of the flavoproteins involved in nucleus-mitochondria crosstalk in cancer. We journey through the influence of flavins on cancer cell energetics, the modulation of RFVTs in malignant transformation, the diagnostic and prognostic significance of FADS, and the implications of RFK in drug resistance and apoptosis. This review also underscores the potential of these molecules and processes as targets for novel diagnostic and therapeutic strategies, offering new avenues for the battle against this relentless disease.
Collapse
Affiliation(s)
- Alessia Nisco
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Maria Tolomeo
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy; Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Katia Zanier
- Biotechnology and Cell Signaling (CNRS/Université de Strasbourg, UMR 7242), Ecole Superieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Maria Barile
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy.
| |
Collapse
|
8
|
Yang Y, Li Q, Chen J, Guo Y, Cai Y, Zhao W, Su S, Sang A. A cuproptosis-related prognostic signature for guiding clinical diagnosis and treatment in uveal melanoma patients. Heliyon 2024; 10:e36324. [PMID: 39247274 PMCID: PMC11378888 DOI: 10.1016/j.heliyon.2024.e36324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024] Open
Abstract
Background Cuproptosis, one of the most recently discovered forms of cell death, is induced by the disruption of copper binding to the mitochondrial respiratory acylation components. However, the mechanism underlying cuproptosis in uveal melanoma (UM) has not yet been adequately studied. Methods RNA and clinical data were obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed cuproptosis-related genes were identified by R software. A prognostic signature was constructed by applying LASSO regression and Cox regression models. The associations between the signature and the immune microenvironment, overall survival, and drug sensitivity were studied. In addition, qPCR and Western blotting were performed on UM cells and RPE cell lines to verify the expression levels of the genes encoding dihydrolipoamide dehydrogenase (DLD) and dihydrolipoamide S-succinyltransferase (DLST) in UM cases. Results Using a cuproptosis-related prognostic signature, UM samples were classified into high- and low-risk groups. A significant difference in overall survival between the two risk groups was evident. Receiver operating characteristic curves demonstrated that the signature is a reliable predictor of prognosis. Immune cell infiltration, drug sensitivity, and immune checkpoint expression were analysed. Significant immune difference between the two high-risk groups was found, and the high expression of immune checkpoints in high-risk groups suggests significant immunotherapy potential. In addition, drug sensitivity analysis experiments suggest that erlotinib may be a potential treatment for high-risk patients. The results of in vitro experiments confirmed that DLD and DLST had higher expression levels in UM cell lines. Conclusions The prognostic signature developed in this study is a reliable biomarker for predicting the prognosis of UM and may serve as a tool for personalised treatment of patients with UM.
Collapse
Affiliation(s)
- Ying Yang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Qixuan Li
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jia Chen
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Yangchen Guo
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yu Cai
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Wenmin Zhao
- Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Shu Su
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Aimin Sang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| |
Collapse
|
9
|
Chen B, Liu J. Mechanisms associated with cuproptosis and implications for ovarian cancer. J Inorg Biochem 2024; 257:112578. [PMID: 38797108 DOI: 10.1016/j.jinorgbio.2024.112578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/08/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024]
Abstract
Ovarian cancer, a profoundly fatal gynecologic neoplasm, exerts a substantial economic strain on nations globally. The formidable challenge of its frequent relapse necessitates the exploration of novel cytotoxic agents, efficacious antineoplastic medications with minimal adverse effects, and strategies to surmount resistance to primary chemotherapeutic agents. These endeavors aim to supplement extant pharmacological interventions and elucidate molecular mechanisms underlying induced cytotoxicity, distinct from conventional therapeutic modalities. Recent scientific research has unveiled a novel form of cellular demise, known as copper-death, which is contingent upon the intracellular concentration of copper. Diverging from conventional mechanisms of cellular demise, copper-death exhibits a pronounced reliance on mitochondrial respiration, particularly the tricarboxylic acid (TCA) cycle. Tumor cells manifest distinctive metabolic profiles and elevated copper levels in comparison to their normal counterparts. The advent of copper-death presents alluring possibilities for targeted therapeutic interventions within the realm of cancer treatment. Hence, the primary objective of this review is to present an overview of the proteins and intricate mechanisms associated with copper-induced cell death, while providing a comprehensive summary of the knowledge acquired regarding potential therapeutic approaches for ovarian cancer. These findings will serve as valuable references to facilitate the advancement of customized therapeutic interventions for ovarian cancer.
Collapse
Affiliation(s)
- Biqing Chen
- The Second Hospital of Jilin University, Changchun, China
| | - Jiaqi Liu
- The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
10
|
Lin Y, Yuan M, Wang G. Copper homeostasis and cuproptosis in gynecological disorders: Pathogenic insights and therapeutic implications. J Trace Elem Med Biol 2024; 84:127436. [PMID: 38547725 DOI: 10.1016/j.jtemb.2024.127436] [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: 12/16/2023] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 05/27/2024]
Abstract
This review comprehensively explores the complex role of copper homeostasis in female reproductive system diseases. As an essential trace element, copper plays a crucial role in various biological functions. Its dysregulation is increasingly recognized as a pivotal factor in the pathogenesis of gynecological disorders. We investigate how copper impacts these diseases, focusing on aspects like oxidative stress, inflammatory responses, immune function, estrogen levels, and angiogenesis. The review highlights significant changes in copper levels in diseases such as cervical, ovarian, endometrial cancer, and endometriosis, underscoring their potential roles in disease mechanisms and therapeutic exploration. The recent discovery of 'cuproptosis,' a novel cell death mechanism induced by copper ions, offers a fresh molecular perspective in understanding these diseases. The review also examines genes associated with cuproptosis, particularly those related to drug resistance, suggesting new strategies to enhance traditional therapy effectiveness. Additionally, we critically evaluate current therapeutic approaches targeting copper homeostasis, including copper ionophores, chelators, and nanoparticles, emphasizing their emerging potential in gynecological disease treatment. This article aims to provide a comprehensive overview of copper's role in female reproductive health, setting the stage for future research to elucidate its mechanisms and develop targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ying Lin
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, China; Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China; Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Disease, Jinan, Shandong Province China; Gynecology Laboratory, Shandong Provincial Hospital, Jinan Shandong Province, China; Gynecology Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan Shandong Province, China
| | - Ming Yuan
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, China; Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Disease, Jinan, Shandong Province China; Gynecology Laboratory, Shandong Provincial Hospital, Jinan Shandong Province, China; Gynecology Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan Shandong Province, China
| | - Guoyun Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, China; Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Disease, Jinan, Shandong Province China; Gynecology Laboratory, Shandong Provincial Hospital, Jinan Shandong Province, China; Gynecology Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan Shandong Province, China.
| |
Collapse
|
11
|
Li Q, Wang T, Zhou Y, Shi J. Cuproptosis in lung cancer: mechanisms and therapeutic potential. Mol Cell Biochem 2024; 479:1487-1499. [PMID: 37480450 DOI: 10.1007/s11010-023-04815-y] [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: 05/03/2023] [Accepted: 07/15/2023] [Indexed: 07/24/2023]
Abstract
Cuproptosis, a recently identified form of cell death that differs from other forms, is induced by the disruption of the binding of copper to mitochondrial respiratory acylation components. Inducing cell cuproptosis and targeting cell copper death pathways are considered potential directions for treating tumor diseases. We have provided a detailed introduction to the metabolic process of copper. In addition, this study attempts to clarify and summarize the relationships between cuproptosis and therapeutic targets and signaling pathways of lung cancer. This review aims to summarize the theoretical achievements for translating the results of lung cancer and cuproptosis experiments into clinical treatment.
Collapse
Affiliation(s)
- Qixuan Li
- Medical School of Nantong University, Nantong University, Nantong, 226001, China
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Tianyi Wang
- Medical School of Nantong University, Nantong University, Nantong, 226001, China
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Youlang Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jiahai Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- School of Public Health, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
12
|
Springer C, Humayun D, Skouta R. Cuproptosis: Unraveling the Mechanisms of Copper-Induced Cell Death and Its Implication in Cancer Therapy. Cancers (Basel) 2024; 16:647. [PMID: 38339398 PMCID: PMC10854864 DOI: 10.3390/cancers16030647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Copper, an essential element for various biological processes, demands precise regulation to avert detrimental health effects and potential cell toxicity. This paper explores the mechanisms of copper-induced cell death, known as cuproptosis, and its potential health and disease implications, including cancer therapy. Copper ionophores, such as elesclomol and disulfiram, increase intracellular copper levels. This elevation triggers oxidative stress and subsequent cell death, offering potential implications in cancer therapy. Additionally, copper ionophores disrupt mitochondrial respiration and protein lipoylation, further contributing to copper toxicity and cell death. Potential targets and biomarkers are identified, as copper can be targeted to those proteins to trigger cuproptosis. The role of copper in different cancers is discussed to understand targeted cancer therapies using copper nanomaterials, copper ionophores, and copper chelators. Furthermore, the role of copper is explored through diseases such as Wilson and Menkes disease to understand the physiological mechanisms of copper. Exploring cuproptosis presents an opportunity to improve treatments for copper-related disorders and various cancers, with the potential to bring significant advancements to modern medicine.
Collapse
Affiliation(s)
- Chloe Springer
- Department of Biology, University of Massachusetts, Amherst, MA 01003, USA;
| | - Danish Humayun
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA;
| | - Rachid Skouta
- Department of Biology, University of Massachusetts, Amherst, MA 01003, USA;
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA;
| |
Collapse
|
13
|
Yan LJ, Wang Y. Roles of Dihydrolipoamide Dehydrogenase in Health and Disease. Antioxid Redox Signal 2023; 39:794-806. [PMID: 37276180 PMCID: PMC10615065 DOI: 10.1089/ars.2022.0181] [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: 10/31/2022] [Revised: 05/22/2023] [Accepted: 05/28/2023] [Indexed: 06/07/2023]
Abstract
Significance: Dihydrolipoamide dehydrogenase (DLDH) is a flavin-dependent disulfide oxidoreductase. The active form of DLDH is a stable homodimer, and its deficiencies have been linked to numerous metabolic disorders. A better understanding of redox and nonredox features of DLDH may reveal druggable targets for disease interventions or preventions. Recent Advances: In this article, the authors review the different roles of DLDH in selected pathological conditions, including its deficiency in humans, its role in stroke and neuroprotection, skin photoaging, Alzheimer's disease, and DLDH as a nondehydrogenating protein, and construction of genetically modified DLDH animal models for further studying the role of DLDH in specific pathological conditions. DLDH is also vulnerable to oxidative modifications in pathological conditions. Critical Issues: Novel animal models need to be constructed using gene knockdown techniques to investigate the redox- and nonredox roles of DLDH in related metabolic diseases. Specific small-molecule DLDH inhibitors need to be discovered. The relationship between modifications of specific amino acid residues in DLDH and given pathological conditions is an interesting area that remains to be comprehensively evaluated. Future Directions: Cell-specific or tissue-specific knockdown of DLDH creating specific pathological conditions will provide more insights into the mechanisms, whereby DLDH may have therapeutic values under a variety of pathological conditions. Antioxid. Redox Signal. 39, 794-806.
Collapse
Affiliation(s)
- Liang-Jun Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Yucheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|