1
|
Wang Y, Li D, Xu K, Wang G, Zhang F. Copper homeostasis and neurodegenerative diseases. Neural Regen Res 2025; 20:3124-3143. [PMID: 39589160 PMCID: PMC11881714 DOI: 10.4103/nrr.nrr-d-24-00642] [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: 07/27/2024] [Accepted: 10/14/2024] [Indexed: 11/27/2024] Open
Abstract
Copper, one of the most prolific transition metals in the body, is required for normal brain physiological activity and allows various functions to work normally through its range of concentrations. Copper homeostasis is meticulously maintained through a complex network of copper-dependent proteins, including copper transporters (CTR1 and CTR2), the two copper ion transporters the Cu -transporting ATPase 1 (ATP7A) and Cu-transporting beta (ATP7B), and the three copper chaperones ATOX1, CCS, and COX17. Disruptions in copper homeostasis can lead to either the deficiency or accumulation of copper in brain tissue. Emerging evidence suggests that abnormal copper metabolism or copper binding to various proteins, including ceruloplasmin and metallothionein, is involved in the pathogenesis of neurodegenerative disorders. However, the exact mechanisms underlying these processes are not known. Copper is a potent oxidant that increases reactive oxygen species production and promotes oxidative stress. Elevated reactive oxygen species levels may further compromise mitochondrial integrity and cause mitochondrial dysfunction. Reactive oxygen species serve as key signaling molecules in copper-induced neuroinflammation, with elevated levels activating several critical inflammatory pathways. Additionally, copper can bind aberrantly to several neuronal proteins, including alpha-synuclein, tau, superoxide dismutase 1, and huntingtin, thereby inducing neurotoxicity and ultimately cell death. This study focuses on the latest literature evaluating the role of copper in neurodegenerative diseases, with a particular focus on copper-containing metalloenzymes and copper-binding proteins in the regulation of copper homeostasis and their involvement in neurodegenerative disease pathogenesis. By synthesizing the current findings on the functions of copper in oxidative stress, neuroinflammation, mitochondrial dysfunction, and protein misfolding, we aim to elucidate the mechanisms by which copper contributes to a wide range of hereditary and neuronal disorders, such as Wilson's disease, Menkes' disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. Potential clinically significant therapeutic targets, including superoxide dismutase 1, D-penicillamine, and 5,7-dichloro-2-[(dimethylamino)methyl]-8-hydroxyquinoline, along with their associated therapeutic agents, are further discussed. Ultimately, we collate evidence that copper homeostasis may function in the underlying etiology of several neurodegenerative diseases and offer novel insights into the potential prevention and treatment of these diseases based on copper homeostasis.
Collapse
Affiliation(s)
- Yuanyuan Wang
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Daidi Li
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Kaifei Xu
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Guoqing Wang
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Feng Zhang
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
2
|
Wang H, Yang Y, Du J. Cuproptosis: the mechanisms of copper-induced cell death and its implication in colorectal cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04263-z. [PMID: 40397118 DOI: 10.1007/s00210-025-04263-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 05/02/2025] [Indexed: 05/22/2025]
Abstract
Colorectal cancer (CRC) represents a prevalent neoplastic disorder of the digestive tract, characterized by elevated incidence and mortality rates. The cellular metabolism of copper within CRC cells is frequently dysregulated, indicating that modifications to copper concentrations may induce cell death and potentially enhance the overall suppression of tumor progression. Cuproptosis, a recently identified form of cellular death, occurs when copper ions bind directly to the lipoylated components of the citric acid cycle (CAC) within mitochondrial respiration, thereby disrupting the balance of iron-sulfur cluster (Fe-S cluster) proteins and ultimately leading to protein toxic stress. The defining traits of cuproptosis include dependence on Cu2+ concentrations and pronounced expression in cells engaged in mitochondrial respiration. This novel mechanism has attracted significant interest within the cancer research community due to its substantial therapeutic potential in oncology. Treatments based on copper demonstrate an inhibitory effect on tumor proliferation and may facilitate approaches for treating tumors that are resistant to conventional chemotherapy. This article aims to review the significance of cuproptosis in CRC, positing that it may serve as a promising strategy for antitumor therapy and an innovative treatment paradigm to address drug resistance in cancer.
Collapse
Affiliation(s)
- Hongyu Wang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Yawen Yang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Juan Du
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China.
| |
Collapse
|
3
|
Pang Z. Copper metabolism in hepatocellular carcinoma: from molecular mechanisms to therapeutic opportunities. Front Mol Biosci 2025; 12:1578693. [PMID: 40433591 PMCID: PMC12106024 DOI: 10.3389/fmolb.2025.1578693] [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: 02/19/2025] [Accepted: 04/17/2025] [Indexed: 05/29/2025] Open
Abstract
Copper is a vital trace metal that facilitates cell proliferation, angiogenesis, and tumour spread. The liver is essential for copper metabolism, hence regulating copper levels is crucial for hepatic health. Hepatocellular carcinoma is a primary liver cancer characterised by a high death rate, and extensive research has shown the substantial impact of copper on its progression. This research primarily examines the molecular mechanisms involved, summarises the regulation of copper homeostasis, and addresses the role of copper metabolism in the promotion and inhibition of hepatocellular carcinoma development. Furthermore, it investigates prospective clinical approaches for targeting copper in the treatment of this disease, intending to establish a theoretical basis for the clinical use of copper in the management of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ziling Pang
- Department of Nursing, School of Medicine, Shihezi University, Shihezi, China
| |
Collapse
|
4
|
Guo Z, Chen D, Yao L, Sun Y, Li D, Le J, Dian Y, Zeng F, Chen X, Deng G. The molecular mechanism and therapeutic landscape of copper and cuproptosis in cancer. Signal Transduct Target Ther 2025; 10:149. [PMID: 40341098 PMCID: PMC12062509 DOI: 10.1038/s41392-025-02192-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/13/2024] [Accepted: 02/17/2025] [Indexed: 05/10/2025] Open
Abstract
Copper, an essential micronutrient, plays significant roles in numerous biological functions. Recent studies have identified imbalances in copper homeostasis across various cancers, along with the emergence of cuproptosis, a novel copper-dependent form of cell death that is crucial for tumor suppression and therapeutic resistance. As a result, manipulating copper levels has garnered increasing interest as an innovative approach to cancer therapy. In this review, we first delineate copper homeostasis at both cellular and systemic levels, clarifying copper's protumorigenic and antitumorigenic functions in cancer. We then outline the key milestones and molecular mechanisms of cuproptosis, including both mitochondria-dependent and independent pathways. Next, we explore the roles of cuproptosis in cancer biology, as well as the interactions mediated by cuproptosis between cancer cells and the immune system. We also summarize emerging therapeutic opportunities targeting copper and discuss the clinical associations of cuproptosis-related genes. Finally, we examine potential biomarkers for cuproptosis and put forward the existing challenges and future prospects for leveraging cuproptosis in cancer therapy. Overall, this review enhances our understanding of the molecular mechanisms and therapeutic landscape of copper and cuproptosis in cancer, highlighting the potential of copper- or cuproptosis-based therapies for cancer treatment.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Danyao Chen
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Jiayuan Le
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| |
Collapse
|
5
|
Xu L, Cao X, Deng Y, Zhang B, Li X, Liu W, Ren W, Tang X, Kong X, Zhang D. Cuproptosis-related genes and agents: implications in tumor drug resistance and future perspectives. Front Pharmacol 2025; 16:1559236. [PMID: 40406488 PMCID: PMC12095339 DOI: 10.3389/fphar.2025.1559236] [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: 01/12/2025] [Accepted: 04/23/2025] [Indexed: 05/26/2025] Open
Abstract
In the field of tumor treatment, drug resistance remains a significant challenge requiring urgent intervention. Recent developments in cell death research have highlighted cuproptosis, a mechanism of cell death induced by copper, as a promising avenue for understanding tumor biology and addressing drug resistance. Cuproptosis is initiated by the dysregulation of copper homeostasis, which in turn triggers mitochondrial metabolic disruptions and induces proteotoxic stress. This process specifically entails the accumulation of lipoylated proteins and the depletion of iron-sulfur cluster proteins within the context of the tricarboxylic acid cycle. Simultaneously, it is accompanied by the activation of distinct signaling pathways that collectively lead to cell death. Emerging evidence highlights the critical role of cuproptosis in addressing tumor drug resistance. However, the core molecular mechanisms of cuproptosis, regulation of the tumor microenvironment, and clinical translation pathways still require further exploration. This review examines the intersection of cuproptosis and tumor drug resistance, detailing the essential roles of cuproptosis-related genes and exploring the therapeutic potential of copper ionophores, chelators, and nanodelivery systems. These mechanisms offer promise for overcoming resistance and advancing tumor precision medicine. By elucidating the molecular mechanisms underlying cuproptosis, this study aims to identify novel therapeutic strategies and targets, thereby paving the way for the development of innovative anti-cancer drugs.
Collapse
Affiliation(s)
- Lingwen Xu
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Xiaolan Cao
- Department of Radiotherapy, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| | - Yuxiao Deng
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Bin Zhang
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Xinzhi Li
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Wentao Liu
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Wenjie Ren
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Xuan Tang
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Xiangyu Kong
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Daizhou Zhang
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| |
Collapse
|
6
|
Lu K, Wijaya CS, Yao Q, Jin H, Feng L. Cuproplasia and cuproptosis, two sides of the coin. Cancer Commun (Lond) 2025; 45:505-524. [PMID: 39865459 PMCID: PMC12067395 DOI: 10.1002/cac2.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/03/2025] [Accepted: 01/19/2025] [Indexed: 01/28/2025] Open
Abstract
Copper is an essential micronutrient in the human body, mainly acting as a crucial cofactor required for a wide range of physiological processes across nearly all cell types. Recent advances revealed that tumor cells seize copper to fulfill their rapid proliferation, metastasis, immune evasion, and so on by reprogramming the copper regulatory network, defined as cuproplasia. Thus, targeting copper chelation to reduce copper levels has been considered a rational tumor therapy strategy. However, overloaded copper ions could be toxic, which leads to the aggregation of lipoylated mitochondrial proteins and the depletion of iron-sulfur clusters, ultimately resulting in cell death, termed cuproptosis. Upon its discovery, cuproptosis has attracted great interest from oncologists, and targeting cuproptosis by copper ionophores exhibits as a potential anti-tumor therapy. In this review, we present the underlying mechanisms involved in cuproplasia and cuproptosis. Additionally, we sum up the chemicals targeting either cuproplasia or cuproptosis for cancer therapy. Further attention should be paid to distinguishing cancer patients who are suitable for targeting cuproplasia or cuproptosis.
Collapse
Affiliation(s)
- Kaizhong Lu
- Department of Medical OncologyZhejiang Key Laboratory of Multi‐omics Precision Diagnosis and Treatment of Liver DiseasesCancer Center of Zhejiang UniversitySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Chandra Sugiarto Wijaya
- Department of Medical OncologyZhejiang Key Laboratory of Multi‐omics Precision Diagnosis and Treatment of Liver DiseasesCancer Center of Zhejiang UniversitySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qinghua Yao
- Department of OncologyThe Second Affiliated Hospital of Zhejiang Chinese Medical UniversityXinhua Hospital of Zhejiang ProvinceHangzhouZhejiangP. R. China
- Key Laboratory for Research on the Pathogenesis of Inflammation‐Cancer Transformation in Intestinal DiseasesZhejiang Engineering Research Center of Intelligent Equipment of Chronic Chinese and Western MedicineHangzhouZhejiangP. R. China
| | - Hongchuan Jin
- Department of Medical OncologyZhejiang Key Laboratory of Multi‐omics Precision Diagnosis and Treatment of Liver DiseasesCancer Center of Zhejiang UniversitySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Lifeng Feng
- Department of Medical OncologyZhejiang Key Laboratory of Multi‐omics Precision Diagnosis and Treatment of Liver DiseasesCancer Center of Zhejiang UniversitySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| |
Collapse
|
7
|
Shan D, Song J, Ren Y, Zhang Y, Ba Y, Luo P, Cheng Q, Xu H, Weng S, Zuo A, Liu S, Han X, Deng J, Liu Z. Copper in cancer: friend or foe? Metabolism, dysregulation, and therapeutic opportunities. Cancer Commun (Lond) 2025; 45:577-607. [PMID: 39945125 PMCID: PMC12067407 DOI: 10.1002/cac2.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 05/13/2025] Open
Abstract
Copper, one of the essential nutrients for the human body, acts as an electron relay in multiple pathways due to its redox properties. Both deficiencies and excesses of copper lead to cellular fragility. Therefore, it can manifest pro- and anti-cancer properties in tumors. Therefore, it is crucial to clarify the copper activity within the cell. We have thoughtfully summarized the metabolic activities of copper from a macro and micro perspective. Cuproptosis, as well as other forms of cell death, is directly or indirectly interfered with by Cu2+, causing cancer cell death. Meanwhile, we did pan-cancer analysis of cuproptosis-related genes to further clarify the roles of these genes. In addition, copper has been found to be involved in multiple pathways within the metastasis of cancer cells. Given the complexity of copper's role, we are compelled to ask: is copper a friend or a foe? Up to now, copper has been used in various clinical applications, including protocols for measurement of copper concentration and bioimaging of radioactive 64Cu. But therapeutically it is still a continuation of the old medicine, and new possibilities need to be explored, such as the use of nanomaterials. Some studies have also shown that copper has considerable interventional power in metabolic cancers, which provides the great applications potential of copper therapy in specific cancer types. This paper reviews the dual roles played by cuproptosis in cancer from the new perspectives of oxidative stress, cell death, and tumor metastasis, and points out the value of its application in specific cancer types, summarizes the value of its testing and imaging from the perspective of clinical application as well as the current feasible options for the new use of the old drugs, and emphasizes the prospects for the application of nano-copper.
Collapse
Affiliation(s)
- Dan Shan
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
- University Hospital GalwayNational University of Ireland GalwayGalwayIreland
- Department of Biobehavioral SciencesColumbia UniversityNew YorkUSA
| | - Jinling Song
- Division of PulmonologyDepartment of PediatricsThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yuqing Ren
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Peng Luo
- The Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Quan Cheng
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanP. R. China
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanP. R. China
| | - Jinhai Deng
- Richard Dimbleby Department of Cancer ResearchComprehensive Cancer Centre, Kings College LondonLondonUK
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanP. R. China
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| |
Collapse
|
8
|
Greco V, Lanza V, Tomasello B, Naletova I, Cairns WRL, Sciuto S, Rizzarelli E. Copper Complexes with New Glycyl-l-histidyl-l-lysine-Hyaluronan Conjugates Show Antioxidant Properties and Osteogenic and Angiogenic Synergistic Effects. Bioconjug Chem 2025; 36:662-675. [PMID: 40123442 DOI: 10.1021/acs.bioconjchem.4c00545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
In recent years, hyaluronic acid (HA) and the natural tripeptide glycyl-l-histidyl-l-lysine (GHK), especially its copper(II) complex (GHK-Cu), individually have been shown to exert helpful properties for bone protection and regeneration. However, they are not strong enough to handle oxidative stress, hydrolytic attack, or environmental conditions. Being aware that conjugation chemistry has recently emerged as an appealing approach for generating new molecular entities capable of preserving the molecular integrity of their moieties or delaying their degradation, herein we present the synthesis of conjugates of HA with GHK (GHK-HA), at different loadings of the tripeptide. GHK-HA binds copper(II) ions and potentiates the chemical and biological properties of the two components in in vitro assays. The results highlight copper's role in promoting the expression and release of certain trophic, angiogenic, and osteogenic factors, including brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), as well as bone morphogenetic protein-2 (BMP-2). The protective and regenerative activities of the metal ion are related to the translocation of its intracellular chaperones Copper Chaperone for Superoxide Dismutase (CCS) and Antioxidant-1 (Atox1) to the nucleus where they act as transcription factors.
Collapse
Affiliation(s)
- Valentina Greco
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Valeria Lanza
- Institute of Crystallography, National Council of Research (CNR), Via P. Gaifami 18, 95126 Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Irina Naletova
- Institute of Crystallography, National Council of Research (CNR), Via P. Gaifami 18, 95126 Catania, Italy
| | - Warren R L Cairns
- CNR-Institute of Polar Sciences (CNR-ISP), Via Torino 155, 30172 Venice, Italy
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Enrico Rizzarelli
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
- Institute of Crystallography, National Council of Research (CNR), Via P. Gaifami 18, 95126 Catania, Italy
| |
Collapse
|
9
|
Yao L, Jiang B, Xu D. Strategies to combat cancer drug resistance: focus on copper metabolism and cuproptosis. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:15. [PMID: 40201308 PMCID: PMC11977383 DOI: 10.20517/cdr.2025.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025]
Abstract
Cancer cells often develop tolerance to chemotherapy, targeted therapy, and immunotherapy drugs either before or during treatment. The significant heterogeneity among various tumors poses a critical challenge in modern cancer research, particularly in overcoming drug resistance. Copper, as an essential trace element in the body, participates in various biological processes of diseases, including cancers. The growth of many types of tumor cells exhibits a heightened dependence on copper. Thus, targeting copper metabolism or inducing cuproptosis may be potential ways to overcome cancer drug resistance. Copper chelators have shown potential in overcoming cancer drug resistance by targeting copper-dependent processes in cancer cells. In contrast, copper ionophores, copper-based nanomaterials, and other small molecules have been used to induce copper-dependent cell death (cuproptosis) in cancer cells, including drug-resistant tumor cells. This review summarizes the regulation of copper metabolism and cuproptosis in cancer cells and the role of copper metabolism and cuproptosis in cancer drug resistance, providing ideas for overcoming cancer resistance in the future.
Collapse
Affiliation(s)
- Leyi Yao
- Zhanjiang Institute of Clinical Medicine, Central People’s Hospital of Zhanjiang, Zhanjiang 524033, Guangdong, China
- Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524033, Guangdong, China
- Authors contributed equally
| | - Baoyi Jiang
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, Guangdong, China
- Authors contributed equally
| | - Dacai Xu
- Zhanjiang Institute of Clinical Medicine, Central People’s Hospital of Zhanjiang, Zhanjiang 524033, Guangdong, China
- Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524033, Guangdong, China
| |
Collapse
|
10
|
Makiabadi E, Nasrollahzadeh J, Nakhaeizadeh R, Shahparvari MR, Roohelhami E. Effects of Perioperative Zinc Supplementation on Copper Circulating Levels and Expression of Metallothionein and Copper Antioxidant Chaperone-1 in Leukocytes in Patients Undergoing CABG Surgery. Biol Trace Elem Res 2025; 203:1297-1304. [PMID: 38865064 DOI: 10.1007/s12011-024-04259-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/01/2024] [Indexed: 06/13/2024]
Abstract
The use of zinc supplement may have a negative effect on copper status. The objective of this study was to evaluate the effect of zinc and vitamin E supplementation on copper and zinc biomarkers in patients undergoing coronary artery bypass graft (CABG) surgery. The study was an add-on project to a previously published randomized controlled trial (NCT05402826) on patients undergoing CABG surgery. Patients in the zinc-vitamin E group (n = 40) received oral zinc (120 mg) and vitamin E (1200 international units) 1 day before surgery, followed by 30 mg of zinc and 200 units of vitamin E per day until 21 days after surgery, while those in the control group (n = 38) received placebo. Plasma levels of copper, ceruloplasmin, superoxide dismutase (SOD) activity, as well as leukocyte gene expression of metallothionein 2A (MT2A) and antioxidant protein 1 (ATOX1), were determined 3 and 21 days after surgery. The plasma copper level in the zinc-vitamin E group was significantly lower than the placebo group on the 3rd postoperative day, but no significant between-group differences were observed on day 21. Plasma ceruloplasmin concentration and SOD activity were not different. Relative mRNA expression of leukocyte MT2A was increased at both times (days 3 and 21 in the zinc-vitamin E group compared to placebo, but ATOX1 expression was not affected. Although the plasma copper level was transiently decreased early after surgery in the zinc-vitamin E group, considering the lack of change in other copper biomarkers, it seems that the use of zinc supplements at the dose used in the present study does not have a significant negative effect on the role of intracellular copper.
Collapse
Affiliation(s)
- Elham Makiabadi
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Nasrollahzadeh
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Nakhaeizadeh
- Department of Surgery, School of Medicine, Shafa Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Reza Shahparvari
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Roohelhami
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Zhao Y, Wang R, Hu C, Wang Y, Li Z, Yin D, Tan S. Complanatoside A disrupts copper homeostasis and induces cuproptosis via directly targeting ATOX1 in prostate cancer. Toxicol Appl Pharmacol 2025; 496:117257. [PMID: 39909165 DOI: 10.1016/j.taap.2025.117257] [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: 11/17/2024] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
Complanatoside A (CA), a flavonoid derived from the Chinese medicinal herb Semen Astragali Complanati, exhibits anticancer activity. However, its effects on prostate cancer (PCa) remain unclear. We aimed to elucidate the anti-PCa effects and underlying mechanisms of action of CA, both in vitro and in vivo. MTT, transwell, wound-healing, and flow cytometry assays were used to detect PCa cell growth, invasion, migration, and apoptosis and cell cycle progression after CA treatment, respectively. The target of CA was predicted to be antioxidant 1 copper chaperone (ATOX1), and its expression levels were determined using immunoblotting analysis. As ATOX1 acts as copper carrier to maintain copper homeostasis and disrupted copper homeostasis leads to oxidative stress in mitochondria and cuproptosis, the concentration of copper, ATP,pyruvate, α-ketoglutamic acid, malondialdehyde, and glutathione were determined and markers of cuproptosis were analyzed by immunoblotting analysis. The copper chelator tetrathiomolybdate was used to identify CA-induced cuproptosis of PCa cells. Finally, a subcutaneous mouse model was constructed, and tumor growth, oxidative stress, and carcinogenesis were analyzed in vivo. Our investigation revealed that CA inhibited PCa cell growth, invasion, migration, and cell cycle progression and induced apoptosis. ATOX1 was downregulated by CA and promoted Cu ion accumulation, which inhibited mitochondrial activity and induced cuproptosis in PCa cells. In addition, CA markedly suppressed tumor growth in vivo and induced cuproptosis in tumor tissues. In conclusion, CA exerts its anti-PCa effects by reducing ATOX1 protein levels and promoting Cu ion accumulation, which in turn, inhibits mitochondrial activity and induces cuproptosis.
Collapse
Affiliation(s)
- Yi Zhao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Ruonan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Chaoyu Hu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Yan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Zengrui Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China; Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Anhui 230012, China.
| | - Song Tan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China.
| |
Collapse
|
12
|
Srishti K, Negi O, Hota PK. Recent Development on Copper-Sensor and its Biological Applications: A Review. J Fluoresc 2025; 35:1273-1313. [PMID: 38416283 DOI: 10.1007/s10895-024-03587-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/14/2024] [Indexed: 02/29/2024]
Abstract
Metal ion recognition is one of the most prospective research topics in the field of chemical sensors due to its wide range of clinical, biological and environmental applications. In this context, hydrazones are well known compounds that exhibit metal sensing and several biological properties due to the presence of N=CH- bond. Some of the biological properties includes anti-cancer, anti-tumor, anti-oxidant, anti-microbial activities. Hydrazones are also used as a ligand to detect metal ion as well as to generate metal complexes that exhibit medicinal properties. Thus, in recent years, many attempts were made to develop novel ligands with enhanced metal sensing and medicinal properties. In this review, some of the recent development on the hydrazones and their copper complexes are covered from the last few years from 2015-2023. These includes significance of copper ions, synthesis, biological properties, mechanism and metal sensing properties of some of the copper complexes were discussed.
Collapse
Affiliation(s)
- Km Srishti
- Department of Chemistry, School of Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Oseen Negi
- Department of Chemistry, School of Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Prasanta Kumar Hota
- Department of Chemistry, School of Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India.
| |
Collapse
|
13
|
Wang D, Guan H. Cuproptosis: A new mechanism for anti-tumour therapy. Pathol Res Pract 2025; 266:155790. [PMID: 39729956 DOI: 10.1016/j.prp.2024.155790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 12/29/2024]
Abstract
As an indispensable trace metal element in the organism, copper acts as a key catalytic cofactor in a wide range of biological processes. Copper homeostasis disorders can be caused by either copper excess or deficiency, and copper homeostasis disorders will affect the normal physiological functions of cells and induce cell death through a variety of mechanisms, such as the emerging cuproptosis model. The imbalance of copper homeostasis will lead to the occurrence of cancer, and copper is a key factor in cell signalling, so copper is involved in the development of cancer by promoting cell proliferation, angiogenesis and metastasis, etc. The therapeutic role of Cuproptosis as a hotspot of research in cancer has also attracted much attention. Therefore, this paper comprehensively searches the literature to review the roles and mechanisms of Cuproptosis in the treatment of malignant tumours, aiming to provide new insights into the role and mechanism of Cuproptosis in anti-malignant tumour therapy and present novel ideas and methods.
Collapse
Affiliation(s)
- Dong Wang
- First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Haoran Guan
- First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
14
|
Li Y, Han Y, Shu Q, Kan YK, Wang Z. Cuproptosis and copper as potential mechanisms and intervention targets in Alzheimer's disease. Biomed Pharmacother 2025; 183:117814. [PMID: 39809124 DOI: 10.1016/j.biopha.2025.117814] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025] Open
Abstract
Recently study has found a new form of copper-dependent death called cuproptosis, which differs from apoptosis, ferroptosis, and necrosis. The main process of cuproptosis is copper directly combined with lipid-acetylated proteins in the TCA cycle of mitochondrial response, leading to the aggregation of lipid-acetylated proteins and the loss of Fe-S cluster proteins, resulting in mitochondrial dysfunction, and eventually causing cell death. Previous studies demonstrated that an imbalance in copper homeostasis exacerbates the pathological progression of Alzheimer's disease (AD) through the induction of oxidative stress, inflammatory response, and the accumulation of Aβ deposition and tau protein hyperphosphorylation. However, the underlying mechanisms remains to be elucidated. More importantly, research identifies the role of cuproptosis and further elucidates the underlying molecular mechanisms in AD. This review summarized the effects of copper metabolism on AD pathology, the characteristics and mechanism of cuproptosis and we discuss the significance of cuproptosis in the pathogenesis of AD.
Collapse
Affiliation(s)
- Ying Li
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ying Han
- Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Qi Shu
- Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Ya-Kun Kan
- The First Hospital of China Medical University, Shenyang 110122, China
| | - Zhuo Wang
- Health Sciences Institute of China Medical University, Shenyang 110122, China.
| |
Collapse
|
15
|
Xie J, Su Y, Shang W, Wu Y, He J, Li T, Shen Y, Zhang Y, Tong X, Bian Q. Involvement of copper in cell death and cancer. Apoptosis 2025; 30:488-505. [PMID: 39702813 DOI: 10.1007/s10495-024-02059-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Copper (cu) is an essential micronutrient required for numerous metabolic processes. It plays a crucial role in cellular respiration by participating in the electron transport chain and facilitating numerous biological reactions. Various diseases, including cancer, demonstrate localized elevation of copper levels and/or alterations in the overall distribution of copper. Modulating local or systemic copper levels as a novel therapeutic approach for treating and ameliorating diseases has emerged as a prominent trend in disease management, particularly in the realm of cancer therapy, which is currently under investigation. The objective of this review is to offer a thorough examination of copper metabolism in both physiological and pathological contexts. Specifically, it delves into how copper ions can effectively target and stimulate tumor cell death via the process known as cuproptosis in cancer patients. Furthermore, this review explores the utilization of three categories of anticancer medications (copper ion carriers, copper complexes, and copper chelating agents) pertaining to copper metabolism within the realm of cancer therapy, elucidating on the distinct mechanisms through which they exert their effects.
Collapse
Affiliation(s)
- Jiahao Xie
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University (Zhejiang Provincial People's Hospital), Hangzhou, Zhejiang, 310014, China
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yue Su
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University (Zhejiang Provincial People's Hospital), Hangzhou, Zhejiang, 310014, China
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wenzhong Shang
- Department of Hematology, Hangzhou First People 's Hospital, Hangzhou, Zhejiang, 310014, China
| | - Yanfang Wu
- Department of Hematology, First People 's Hospital of Fuyang District, Hangzhou, Zhejiang, 310014, China
| | - Junjia He
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Ting Li
- College of Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yeyu Shen
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Youni Zhang
- Department of Laboratory Medicine, Tiantai People's Hospital, Taizhou, Zhejiang Province, 317200, China.
| | - Xiangmin Tong
- Department of Hematology, First People 's Hospital of Fuyang District, Hangzhou, Zhejiang, 310014, China.
| | - Qiong Bian
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
16
|
Li SR, Tao SY, Li Q, Hu CY, Sun ZJ. Harnessing nanomaterials for copper-induced cell death. Biomaterials 2025; 313:122805. [PMID: 39250865 DOI: 10.1016/j.biomaterials.2024.122805] [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: 03/12/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/11/2024]
Abstract
Copper (Cu), an essential micronutrient with redox properties, plays a pivotal role in a wide array of pathological and physiological processes across virtually all cell types. Maintaining an optimal copper concentration is critical for cellular survival: insufficient copper levels disrupt respiration and metabolism, while excess copper compromises cell viability, potentially leading to cell death. Similarly, in the context of cancer, copper exhibits a dual role: appropriate amount of copper can promote tumor progression and be an accomplice, yet beyond befitting level, copper can bring about multiple types of cell death, including autophagy, apoptosis, ferroptosis, immunogenic cell death, pyroptosis, and cuproptosis. These forms of cell death are beneficial against cancer progression; however, achieving precise copper regulation within tumors remains a significant challenge in the pursuit of effective cancer therapies. The emergence of nanodrug delivery systems, distinguished by their precise targeting, controlled release, high payload capacity, and the ability to co-deliver multiple agents, has revitalized interest in exploiting copper's precise regulatory capabilities. Nevertheless, there remains a dearth of comprehensive review of copper's bidirectional effects on tumorigenesis and the role of copper-based nanomaterials in modulating tumor progression. This paper aims to address this gap by elucidating the complex role in cancer biology and highlighting its potential as a therapeutic target. Through an exploration of copper's dualistic nature and the application of nanotechnology, this review seeks to offer novel insights and guide future research in advancing cancer treatment.
Collapse
Affiliation(s)
- Su-Ran Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China
| | - Shi-Yue Tao
- Bathune School of Stomatology, Jilin University, Changchun, 130021, Jilin, PR China
| | - Qian Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China
| | - Chuan-Yu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China.
| |
Collapse
|
17
|
Yu HF, Zeng QR, Xiao P, Yang D, Ping Y, Liu M, Yu Z, Wang C, Teng CB. Hippo-YAP signaling alleviates copper-induced mitochondrial dysfunction and oxidative damage via the ATOX1-PPA2 pathway. Int J Biol Macromol 2025; 290:138908. [PMID: 39706439 DOI: 10.1016/j.ijbiomac.2024.138908] [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: 07/16/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Hippo signaling plays a crucial role in the cellular response to various stressors, such as mechanical stress, metabolic stress, and hypoxic stress. However, its physiological significance in copper (Cu) stress remains poorly understood. Here, we demonstrated aberrant activation of Hippo-YAP signaling in sheep pancreas and pancreatic organoids exposed to excessive Cu, accompanied by significant pathological changes, elevated levels of oxidative stress, and impaired mitochondrial structure and function. The inhibition of Hippo signaling or overexpression of YAP protected against Cu-induced damage by improving mitochondrial function and maintaining cellular Cu homeostasis. YAP interacted with TEAD and upregulated the expression of Cu chaperone ATOX1, a key regulator of intracellular Cu homeostasis. ATOX1 restored mitochondrial function under Cu stress by reducing mitochondrial superoxide levels, increasing ATP production and mitochondrial membrane potential. Additionally, our findings confirmed that ATOX1 indirectly bound to the PPA2 promoter and increased its transcription. Notably, the restoration of ATP production in mitochondria mediated by PPA2 overexpression facilitated efficient intracellular Cu efflux, allowing rapid and precise reestablishment of intracellular Cu homeostasis under Cu stress. Collectively, Hippo-YAP signaling alleviates Cu-induced oxidative damage by restoring mitochondrial function through activation of PPA2 transcription depending on ATOX1, thereby ensuring cellular Cu efflux and enhancing antioxidant capacity.
Collapse
Affiliation(s)
- Hai-Fan Yu
- Laboratory of Animal Developmental Biology, College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Qi-Ran Zeng
- Laboratory of Animal Developmental Biology, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Pengyu Xiao
- Laboratory of Animal Developmental Biology, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Dian Yang
- Laboratory of Animal Developmental Biology, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Yue Ping
- Laboratory of Animal Developmental Biology, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Miao Liu
- Laboratory of Animal Developmental Biology, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Ze Yu
- Laboratory of Animal Developmental Biology, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Chunsheng Wang
- Laboratory of Animal Developmental Biology, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Chun-Bo Teng
- Laboratory of Animal Developmental Biology, College of Life Science, Northeast Forestry University, Harbin 150040, China.
| |
Collapse
|
18
|
Liu Z, Gan Y, Shen Z, Cai S, Wang X, Li Y, Li X, Fu H, Chen J, Li N. Role of copper homeostasis and cuproptosis in heart failure pathogenesis: implications for therapeutic strategies. Front Pharmacol 2025; 15:1527901. [PMID: 39850564 PMCID: PMC11754225 DOI: 10.3389/fphar.2024.1527901] [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: 11/14/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Copper is an essential micronutrient involved in various physiological processes in various cell types. Consequently, dysregulation of copper homeostasis-either excessive or deficient-can lead to pathological changes, such as heart failure (HF). Recently, a new type of copper-dependent cell death known as cuproptosis has drawn increasing attention to the impact of copper dyshomeostasis on HF. Notably, copper dyshomeostasis was associated with the occurrence of HF. Hence, this review aimed to investigate the biological processes involved in copper uptake, transport, excretion, and storage at both the cellular and systemic levels in terms of cuproptosis and HF, along with the underlying mechanisms of action. Additionally, the role of cuproptosis and its related mitochondrial dysfunction in HF pathogenesis was analyzed. Finally, we reviewed the therapeutic potential of current drugs that target copper metabolism for treating HF. Overall, the conclusions of this review revealed the therapeutic potential of copper-based therapies that target cuproptosis for the development of strategies for the treatment of HF.
Collapse
Affiliation(s)
- Zhichao Liu
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Yongkang Gan
- Department of Vascular Surgery, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Zhen Shen
- Department of Clinical Laboratory, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Siqi Cai
- College of Art, Nanjing University of Information Science and Technology, Nanjing, Jiangsu, China
| | - Xizhen Wang
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Yong Li
- Experimental Center for Medical Research, Shandong Second Medical University, Weifang, Shandong, China
| | - Xiaofeng Li
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huanjie Fu
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinhong Chen
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Ningcen Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
19
|
Liu X, Zhang W, Wei S, Liang X, Luo B. Targeting cuproptosis with nano material: new way to enhancing the efficacy of immunotherapy in colorectal cancer. Front Pharmacol 2024; 15:1451067. [PMID: 39691393 PMCID: PMC11649426 DOI: 10.3389/fphar.2024.1451067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Colorectal cancer has emerged as one of the predominant malignant tumors globally. Immunotherapy, as a novel therapeutic methodology, has opened up new possibilities for colorectal cancer patients. However, its actual clinical efficacy requires further enhancement. Copper, as an exceptionally crucial trace element, can influence various signaling pathways, gene expression, and biological metabolic processes in cells, thus playing a critical role in the pathogenesis of colorectal cancer. Recent studies have revealed that cuproptosis, a novel mode of cell death, holds promise to become a potential target to overcome resistance to colorectal cancer immunotherapy. This shows substantial potential in the combination treatment of colorectal cancer. Conveying copper into tumor cells via a nano-drug delivery system to induce cuproptosis of colorectal cancer cells could offer a potential strategy for eliminating drug-resistant colorectal cancer cells and vastly improving the efficacy of immunotherapy while ultimately destroy colorectal tumors. Moreover, combining the cuproptosis induction strategy with other anti-tumor approaches such as photothermal therapy, photodynamic therapy, and chemodynamic therapy could further enhance its therapeutic effect. This review aims to illuminate the practical significance of cuproptosis and cuproptosis-inducing nano-drugs in colorectal cancer immunotherapy, and scrutinize the current challenges and limitations of this methodology, thereby providing innovative thoughts and references for the advancement of cuproptosis-based colorectal cancer immunotherapy strategies.
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, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| | - Wanqiu Zhang
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| | - Shaozhong Wei
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinjun Liang
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
- Department of Abdominal Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 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, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| |
Collapse
|
20
|
Li J, Peng C, Huang C, Wan L, Wang K, Wu P, Chen T, Sun G, Guo R, Lin H, Ji Z. Metal Ruthenium Complexes Treat Spinal Cord Injury By Alleviating Oxidative Stress Through Interaction With Antioxidant 1 Copper Chaperone Protein. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407225. [PMID: 39412068 PMCID: PMC11615763 DOI: 10.1002/advs.202407225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/19/2024] [Indexed: 12/06/2024]
Abstract
Oxidative stress is a major factor affecting spinal cord injury (SCI) prognosis. A ruthenium metal complex can aid in treating SCI by scavenging reactive oxygen species via a protein-regulated mechanism to alleviate oxidative stress. This study aimed to introduce a pioneering strategy for SCI treatment by designing two novel half-sandwich ruthenium (II) complexes containing diverse N^N-chelating ligands. The general formula is [(η6-Arene)Ru(N^N)Cl]PF6, where arene is either 2-phenylethanol-1-ol (bz-EA) or 3-phenylpropanol-1-ol (bz-PA), and the N^N-chelating ligands are fluorine-based imino-pyridyl ligands. This study shows that these ruthenium metal complexes protect neurons by scavenging reactive oxygen species. Notably, η6-Arene substitution from bz-PA to bz-EA significantly enhances reactive oxygen species scavenging ability and neuroprotective effect. Additionally, molecular dynamics simulations indicate that the ruthenium metal complex increases Antioxidant 1 Copper Chaperone protein expression, reduces oxidative stress, and protects neurons during SCI treatment. Furthermore, ruthenium metal complex protected spinal cord neurons and stimulated their regeneration, which improves electrical signals and motor functions in mice with SCI. Thus, this treatment strategy using ruthenium metal complexes can be a new therapeutic approach for the efficient treatment of SCI.
Collapse
Affiliation(s)
- Juanjuan Li
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
- Department of UrologyGuangzhou Institute of UrologyGuangdong Key Laboratory of Urologythe State Key Laboratory of Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Medical UniversityGuangzhouGuangdong510230China
| | - Cheng Peng
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
| | - Caiqiang Huang
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
| | - Li Wan
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
| | - Ke Wang
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
| | - Ping Wu
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
| | - Tianjun Chen
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
| | - Guodong Sun
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuan517000China
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education InstitutesGuangdong Provincial Engineering and Technological Research Center for Drug Carrier DevelopmentDepartment of Biomedical EngineeringJinan UniversityGuangzhou510632China
| | - Hongsheng Lin
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
| | - Zhisheng Ji
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouGuangdong510632China
| |
Collapse
|
21
|
Verdejo-Torres O, Klein DC, Novoa-Aponte L, Carrazco-Carrillo J, Bonilla-Pinto D, Rivera A, Bakhshian A, Fitisemanu FM, Jiménez-González ML, Flinn L, Pezacki AT, Lanzirotti A, Ortiz Frade LA, Chang CJ, Navea JG, Blaby-Haas CE, Hainer SJ, Padilla-Benavides T. Cysteine Rich Intestinal Protein 2 is a copper-responsive regulator of skeletal muscle differentiation and metal homeostasis. PLoS Genet 2024; 20:e1011495. [PMID: 39637238 DOI: 10.1371/journal.pgen.1011495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/26/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Copper (Cu) is essential for respiration, neurotransmitter synthesis, oxidative stress response, and transcription regulation, with imbalances leading to neurological, cognitive, and muscular disorders. Here we show the role of a novel Cu-binding protein (Cu-BP) in mammalian transcriptional regulation, specifically on skeletal muscle differentiation using murine primary myoblasts. Utilizing synchrotron X-ray fluorescence-mass spectrometry, we identified murine cysteine-rich intestinal protein 2 (mCrip2) as a key Cu-BP abundant in both nuclear and cytosolic fractions. mCrip2 binds two to four Cu+ ions with high affinity and presents limited redox potential. CRISPR/Cas9-mediated deletion of mCrip2 impaired myogenesis, likely due to Cu accumulation in cells. CUT&RUN and transcriptome analyses revealed its association with gene promoters, including MyoD1 and metallothioneins, suggesting a novel Cu-responsive regulatory role for mCrip2. Our work describes the significance of mCrip2 in skeletal muscle differentiation and metal homeostasis, expanding understanding of the Cu-network in myoblasts. Copper (Cu) is essential for various cellular processes, including respiration and stress response, but imbalances can cause serious health issues. This study reveals a new Cu-binding protein (Cu-BP) involved in muscle development in primary myoblasts. Using unbiased metalloproteomic techniques and high throughput sequencing, we identified mCrip2 as a key Cu-BP found in cell nuclei and cytoplasm. mCrip2 binds up to four Cu+ ions and has a limited redox potential. Deleting mCrip2 using CRISPR/Cas9 disrupted muscle formation due to Cu accumulation. Further analyses showed that mCrip2 regulates the expression of genes like MyoD1, essential for muscle differentiation, and metallothioneins in response to copper supplementation. This research highlights the importance of mCrip2 in muscle development and metal homeostasis, providing new insights into the Cu-network in cells.
Collapse
Affiliation(s)
- Odette Verdejo-Torres
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, Connecticut, United States of America
| | - David C Klein
- Department of Biological Sciences. University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Lorena Novoa-Aponte
- Department of Chemistry and Biochemistry. Worcester Polytechnic Institute, Worcester, Massachusetts, United States of America
| | - Jaime Carrazco-Carrillo
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, Connecticut, United States of America
| | - Denzel Bonilla-Pinto
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, Connecticut, United States of America
| | - Antonio Rivera
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, Connecticut, United States of America
| | - Arpie Bakhshian
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, Connecticut, United States of America
| | - Fa'alataitaua M Fitisemanu
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, Connecticut, United States of America
| | - Martha L Jiménez-González
- Departamento de Electroquímica, Centro de Investigación y Desarrollo Tecnológico en Electroquímica, Santiago de Querétaro, Querétaro, México
| | - Lyra Flinn
- Chemistry Department. Skidmore College, Saratoga Springs, New York, United States of America
| | - Aidan T Pezacki
- Department of Chemistry, Princeton University, Princeton, New Jersey, United States of America
- Department of Chemistry. University of California, Berkeley, California, United States of America
| | - Antonio Lanzirotti
- Center for Advanced Radiation Sources, The University of Chicago, Lemont, Illinois, United States of America
| | - Luis Antonio Ortiz Frade
- Departamento de Electroquímica, Centro de Investigación y Desarrollo Tecnológico en Electroquímica, Santiago de Querétaro, Querétaro, México
| | - Christopher J Chang
- Department of Chemistry, Princeton University, Princeton, New Jersey, United States of America
- Department of Chemistry. University of California, Berkeley, California, United States of America
- Department of Molecular and Cell Biology. University of California, Berkeley, California, United States of America
| | - Juan G Navea
- Chemistry Department. Skidmore College, Saratoga Springs, New York, United States of America
| | - Crysten E Blaby-Haas
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, California & DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Sarah J Hainer
- Department of Biological Sciences. University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvanian United States of America
| | - Teresita Padilla-Benavides
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, Connecticut, United States of America
| |
Collapse
|
22
|
Małyszko M, Przybyłkowski A. Copper and Colorectal Cancer. Cancers (Basel) 2024; 16:3691. [PMID: 39518128 PMCID: PMC11544869 DOI: 10.3390/cancers16213691] [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: 09/17/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Minerals constitute only 5% of the typical human diet but are vital for health and functionality. Copper, a trace element, is absorbed by the human gut at 30-40% from diets typical of industrialized countries. The liver produces metallothioneins, which store copper. Copper is crucial for mitochondrial respiration, pigmentation, iron transport, antioxidant defense, hormone production, and extracellular matrix biosynthesis. Copper deficiency, often caused by mutations in the ATP7A gene, results in Menkes disease, an X-linked recessive disorder. On the contrary, Wilson disease is characterized by toxic copper accumulation. Cuproptosis, a unique form of cell death regulated by copper, is a subtype of necrosis induced by enhanced mitochondrial metabolism and intracellular copper accumulation. This process can reduce the malignant potential of tumor cells by inhibiting glucose metabolism. Therapeutically, copper and its complexes have shown efficacy in malignancy treatments. The disruption of copper homeostasis and excessive cuproplasia are significant in colorectal cancer development and metastasis. Therefore, manipulating copper status presents a potential therapeutic target for colorectal cancer, using copper chelators to inhibit copper formation or copper ion carriers to promote cuproptosis. This review highlights the role of copper in human physiology and pathology, emphasizing its impact on colorectal cancer and potential therapeutic strategies. Future AI-based approaches are anticipated to accelerate the development of new compounds targeting cuproptosis and copper disruption in colorectal cancer.
Collapse
Affiliation(s)
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland;
| |
Collapse
|
23
|
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
|
24
|
Noh D, Lee H, Lee S, Sun IC, Yoon HY. Copper-Based Nanomedicines for Cuproptosis-Mediated Effective Cancer Treatment. Biomater Res 2024; 28:0094. [PMID: 39430913 PMCID: PMC11486892 DOI: 10.34133/bmr.0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
The recent discovery of cuproptosis, a novel copper-ion-induced cell death pathway, has suggested the novel therapeutic potential for treating heterogeneous and drug-resistant cancers. Currently, copper ionophore-based therapeutics have been designed to treat cancers, utilizing copper ions as a strategic tool to impede tumor proliferation and promote cellular demise. However, limitations of copper ionophore-based therapies include nontargeted delivery of copper ions, low tumor accumulation, and short half-life. Strategies to enhance specificity involve targeting intracellular cuproptosis mechanisms using nanotechnology-based drugs. Additionally, the importance of exploring combination therapies cannot be overstated, as they are a key strategy in improving the efficacy of cancer treatments. Recent studies have reported the anticancer effects of nanomedicines that can induce cuproptosis of cancer both in vitro and in vivo. These cuproptosis-targeted nanomedicines could improve delivery efficiency with the pharmacokinetic properties of copper ion, resulting in increasing cuproptosis-based anticancer effects. This review will summarize the intricate nexus between copper ion and carcinogenesis, examining the pivotal roles of copper homeostasis and its dysregulation in cancer progression and fatality. Furthermore, we will introduce the latest advances in cuproptosis-targeted nanomedicines for cancer treatment. Finally, the challenges in cuproptosis-based nanomedicines will be discussed for future development directions.
Collapse
Affiliation(s)
- Dahye Noh
- Medicinal Materials Research Center, Biomedical Research Institute,
Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School,
University of Science and Technology (UST), Hwarang-ro14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hokyung Lee
- Medicinal Materials Research Center, Biomedical Research Institute,
Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Department of Fundamental Pharmaceutical Sciences, College of Pharmacy,
Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sangmin Lee
- Department of Fundamental Pharmaceutical Sciences, College of Pharmacy,
Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - In-Cheol Sun
- Medicinal Materials Research Center, Biomedical Research Institute,
Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hong Yeol Yoon
- Medicinal Materials Research Center, Biomedical Research Institute,
Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School,
University of Science and Technology (UST), Hwarang-ro14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
25
|
Fitisemanu FM, Padilla-Benavides T. Emerging perspectives of copper-mediated transcriptional regulation in mammalian cell development. Metallomics 2024; 16:mfae046. [PMID: 39375833 PMCID: PMC11503025 DOI: 10.1093/mtomcs/mfae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Copper (Cu) is a vital micronutrient necessary for proper development and function of mammalian cells and tissues. Cu mediates the function of redox active enzymes that facilitate metabolic processes and signaling pathways. Cu levels are tightly regulated by a network of Cu-binding transporters, chaperones, and small molecule ligands. Extensive research has focused on the mammalian Cu homeostasis (cuprostasis) network and pathologies, which result from mutations and perturbations. There are roles for Cu-binding proteins as transcription factors (Cu-TFs) and regulators that mediate metal homeostasis through the activation or repression of genes associated with Cu handling. Emerging evidence suggests that Cu and some Cu-TFs may be involved in the regulation of targets related to development-expanding the biological roles of Cu-binding proteins. Cu and Cu-TFs are implicated in embryonic and tissue-specific development alongside the mediation of the cellular response to oxidative stress and hypoxia. Cu-TFs are also involved in the regulation of targets implicated in neurological disorders, providing new biomarkers and therapeutic targets for diseases such as Parkinson's disease, prion disease, and Friedreich's ataxia. This review provides a critical analysis of the current understanding of the role of Cu and cuproproteins in transcriptional regulation.
Collapse
|
26
|
Focaccio A, Rossi L, De Luca A. A spotlight on the role of copper in the epithelial to mesenchymal transition. Life Sci 2024; 354:122972. [PMID: 39142503 DOI: 10.1016/j.lfs.2024.122972] [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: 05/01/2024] [Revised: 07/29/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
The complex process known as epithelial to mesenchymal transition (EMT) plays a fundamental role in several biological settings, encompassing embryonic development, wound healing, and pathological conditions such as cancer and fibrosis. In recent years, a bulk of research has brought to light the key role of copper, a trace element with essential functions in cellular metabolism, cancer initiation and progression. Indeed, copper, besides functioning as cofactor of enzymes required for essential cellular processes, such as energy production and oxidation reactions, has emerged as an allosteric regulator of kinases whose activity is required to fulfill cancer dissemination through the EMT. In this comprehensive review, we try to describe the intricate relationship between the transition metal copper and EMT, spanning from the earliest foundational studies to the latest advancements. Our aim is to shed light on the multifaceted roles undertaken by copper in EMT in cancer and to unveil the diverse mechanisms by which copper homeostasis exerts its influence over EMT regulators, signaling pathways, cell metabolic reprogramming and transcription factors ultimately contributing to the spread of cancer. Therefore, this review not only may contribute to a deeper comprehension of copper-mediated mechanisms in EMT but also supports the hypothesis that targeting copper may contribute to counteract the progression of EMT-associated pathologies.
Collapse
Affiliation(s)
- Antonio Focaccio
- PhD School in Cellular and Molecular Biology, Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Luisa Rossi
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Anastasia De Luca
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy.
| |
Collapse
|
27
|
Huang X, Lian M, Li C. Copper homeostasis and cuproptosis in gynecological cancers. Front Cell Dev Biol 2024; 12:1459183. [PMID: 39386020 PMCID: PMC11461353 DOI: 10.3389/fcell.2024.1459183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
Copper (Cu) is an essential trace element involved in a variety of biological processes, such as antioxidant defense, mitochondrial respiration, and bio-compound synthesis. In recent years, a novel theory called cuproptosis has emerged to explain how Cu induces programmed cell death. Cu targets lipoylated enzymes in the tricarboxylic acid cycle and subsequently triggers the oligomerization of lipoylated dihydrolipoamide S-acetyltransferase, leading to the loss of Fe-S clusters and induction of heat shock protein 70. Gynecological malignancies including cervical cancer, ovarian cancer and uterine corpus endometrial carcinoma significantly impact women's quality of life and even pose a threat to their lives. Excessive Cu can promote cancer progression by enhancing tumor growth, proliferation, angiogenesis and metastasis through multiple signaling pathways. However, there are few studies investigating gynecological cancers in relation to cuproptosis. Therefore, this review discusses Cu homeostasis and cuproptosis while exploring the potential use of cuproptosis for prognosis prediction as well as its implications in the progression and treatment of gynecological cancers. Additionally, we explore the application of Cu ionophore therapy in treating gynecological malignancies.
Collapse
Affiliation(s)
- Xiaodi Huang
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Obstetrics and Gynecology, Shenzhen PKU-HKUST Medical Center, Shenzhen, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, China
| | - Mengyi Lian
- Department of Obstetrics and Gynecology, Longquan People’s Hospital, Lishui, China
| | - Changzhong Li
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Obstetrics and Gynecology, Shenzhen PKU-HKUST Medical Center, Shenzhen, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, China
| |
Collapse
|
28
|
Qian J, Zhao L, Xu L, Zhao J, Tang Y, Yu M, Lin J, Ding L, Cui Q. Cell Death: Mechanisms and Potential Targets in Breast Cancer Therapy. Int J Mol Sci 2024; 25:9703. [PMID: 39273650 PMCID: PMC11395276 DOI: 10.3390/ijms25179703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Breast cancer (BC) has become the most life-threatening cancer to women worldwide, with multiple subtypes, poor prognosis, and rising mortality. The molecular heterogeneity of BC limits the efficacy and represents challenges for existing therapies, mainly due to the unpredictable clinical response, the reason for which probably lies in the interactions and alterations of diverse cell death pathways. However, most studies and drugs have focused on a single type of cell death, while the therapeutic opportunities related to other cell death pathways are often neglected. Therefore, it is critical to identify the predominant type of cell death, the transition to different cell death patterns during treatment, and the underlying regulatory mechanisms in BC. In this review, we summarize the characteristics of various forms of cell death, including PANoptosis (pyroptosis, apoptosis, necroptosis), autophagy, ferroptosis, and cuproptosis, and discuss their triggers and signaling cascades in BC, which may provide a reference for future pathogenesis research and allow for the development of novel targeted therapeutics in BC.
Collapse
Affiliation(s)
- Jiangying Qian
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Linna Zhao
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Ling Xu
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Jin Zhao
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Yongxu Tang
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Min Yu
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Jie Lin
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Lei Ding
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Qinghua Cui
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| |
Collapse
|
29
|
Pan C, Ji Z, Wang Q, Zhang Z, Wang Z, Li C, Lu S, Ge P. Cuproptosis: Mechanisms, biological significance, and advances in disease treatment-A systematic review. CNS Neurosci Ther 2024; 30:e70039. [PMID: 39267265 PMCID: PMC11392831 DOI: 10.1111/cns.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Copper is an essential trace element for biological systems, as it plays a critical role in the activity of various enzymes and metabolic processes. However, the dysregulation of copper homeostasis is closely associated with the onset and progression of numerous diseases. In recent years, copper-induced cell death, a novel form of cellular demise, has garnered significant attention. This process is characterized by the abnormal accumulation of intracellular copper ions, leading to cellular dysfunction and eventual cell death. Copper toxicity occurs through the interaction of copper with acylated enzymes in the tricarboxylic acid (TCA) cycle. This interaction results in subsequent protein aggregation, causing proteotoxic stress and ultimately resulting in cell death. Despite the promise of these findings, the detailed mechanisms and broader implications of cuproptosis remain underexplored. Therefore, our study aimed to investigate the role of copper in cell death and autophagy, focusing on the molecular mechanisms of cuproptosis. We also aimed to discuss recent advancements in copper-related research across various diseases and tumors, providing insights for future studies and potential therapeutic applications. MAIN BODY This review delves into the biological significance of copper metabolism and the molecular mechanisms underlying copper-induced cell death. Furthermore, we discuss the role of copper toxicity in the pathogenesis of various diseases, emphasizing recent advancements in the field of oncology. Additionally, we explore the therapeutic potential of targeting copper toxicity. CONCLUSION The study highlights the need for further research to explore alternative pathways of copper-induced cell death, detailed mechanisms of cuproptosis, and biomarkers for copper poisoning. Future research should focus on exploring the molecular mechanisms of cuproptosis, developing new therapeutic strategies, and verifying their safety and efficacy in clinical trials.
Collapse
Affiliation(s)
- Chengliang Pan
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Zhilin Ji
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Qingxuan Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Zhao Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Zhenchuan Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Chen Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Shan Lu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Pengfei Ge
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
30
|
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: 11] [Impact Index Per Article: 11.0] [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
|
31
|
Zhao R, Sukocheva O, Tse E, Neganova M, Aleksandrova Y, Zheng Y, Gu H, Zhao D, Madhunapantula SV, Zhu X, Liu J, Fan R. Cuproptosis, the novel type of oxidation-induced cell death in thoracic cancers: can it enhance the success of immunotherapy? Cell Commun Signal 2024; 22:379. [PMID: 39068453 PMCID: PMC11282696 DOI: 10.1186/s12964-024-01743-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Copper is an important metal micronutrient, required for the balanced growth and normal physiological functions of human organism. Copper-related toxicity and dysbalanced metabolism were associated with the disruption of intracellular respiration and the development of various diseases, including cancer. Notably, copper-induced cell death was defined as cuproptosis which was also observed in malignant cells, representing an attractive anti-cancer instrument. Excess of intracellular copper leads to the aggregation of lipoylation proteins and toxic stress, ultimately resulting in the activation of cell death. Differential expression of cuproptosis-related genes was detected in normal and malignant tissues. Cuproptosis-related genes were also linked to the regulation of oxidative stress, immune cell responses, and composition of tumor microenvironment. Activation of cuproptosis was associated with increased expression of redox-metabolism-regulating genes, such as ferredoxin 1 (FDX1), lipoic acid synthetase (LIAS), lipoyltransferase 1 (LIPT1), dihydrolipoamide dehydrogenase (DLD), drolipoamide S-acetyltransferase (DLAT), pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1), and pyruvate dehydrogenase E1 subunit beta (PDHB)). Accordingly, copper-activated network was suggested as an attractive target in cancer therapy. Mechanisms of cuproptosis and regulation of cuproptosis-related genes in different cancers and tumor microenvironment are discussed in this study. The analysis of current findings indicates that therapeutic regulation of copper signaling, and activation of cuproptosis-related targets may provide an effective tool for the improvement of immunotherapy regimens.
Collapse
Affiliation(s)
- Ruiwen Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Olga Sukocheva
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia.
| | - Edmund Tse
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia
| | - Margarita Neganova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yulia Aleksandrova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yufei Zheng
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Gu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Deyao Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - SabbaRao V Madhunapantula
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Xiaorong Zhu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Junqi Liu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ruitai Fan
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
32
|
Vo TTT, Peng TY, Nguyen TH, Bui TNH, Wang CS, Lee WJ, Chen YL, Wu YC, Lee IT. The crosstalk between copper-induced oxidative stress and cuproptosis: a novel potential anticancer paradigm. Cell Commun Signal 2024; 22:353. [PMID: 38970072 PMCID: PMC11225285 DOI: 10.1186/s12964-024-01726-3] [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: 04/11/2024] [Accepted: 06/25/2024] [Indexed: 07/07/2024] Open
Abstract
Copper is a crucial trace element that plays a role in various pathophysiological processes in the human body. Copper also acts as a transition metal involved in redox reactions, contributing to the generation of reactive oxygen species (ROS). Under prolonged and increased ROS levels, oxidative stress occurs, which has been implicated in different types of regulated cell death. The recent discovery of cuproptosis, a copper-dependent regulated cell death pathway that is distinct from other known regulated cell death forms, has raised interest to researchers in the field of cancer therapy. Herein, the present work aims to outline the current understanding of cuproptosis, with an emphasis on its anticancer activities through the interplay with copper-induced oxidative stress, thereby providing new ideas for therapeutic approaches targeting modes of cell death in the future.
Collapse
Affiliation(s)
- Thi Thuy Tien Vo
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Tzu-Yu Peng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Thi Hong Nguyen
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Trang Ngoc Huyen Bui
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Ching-Shuen Wang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Wei-Ju Lee
- School of Food Safety, College of Nutrition, Taipei Medical University, Taipei, 110301, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Yang-Che Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
| |
Collapse
|
33
|
Huang XY, Shen JY, Huang K, Wang L, Sethi G, Ma Z. Cuproptosis in cancers: Function and implications from bench to bedside. Biomed Pharmacother 2024; 176:116874. [PMID: 38850661 DOI: 10.1016/j.biopha.2024.116874] [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: 03/07/2024] [Revised: 05/15/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Copper, an indispensable micronutrient, is implicated in numerous vital biological processes and is essential for all physiological activities. Recently, the discovery of a novel type of copper-dependent cell death, known as cuproptosis, has shed light on its role in cancer development. Extensive research is currently underway to unravel the mechanisms underlying cuproptosis and its correlation with various cancer types. In this review, we summarize the findings regarding the roles and mechanisms of cuproptosis in various cancer types, including colorectal cancer, lung cancer, gastric cancer, breast cancer, liver cancer and cutaneous melanoma. Furthermore, the effects of copper-related agents such as copper chelators and copper ionophores on cell proliferation, apoptosis, angiogenesis, tumor immunity, and chemotherapy resistance have been explored in cancer preclinical and clinical trials. These insights provide promising avenues for the development of prospective anticancer drugs aimed at inducing cuproptosis.
Collapse
Affiliation(s)
- Xin-Yi Huang
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China
| | - Jia-Yang Shen
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China
| | - Ke Huang
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore; Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore.
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China.
| |
Collapse
|
34
|
Kamiya T. Role of copper and SOD3-mediated extracellular redox regulation in tumor progression. J Clin Biochem Nutr 2024; 75:1-6. [PMID: 39070539 PMCID: PMC11273271 DOI: 10.3164/jcbn.24-14] [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: 01/22/2024] [Accepted: 04/03/2024] [Indexed: 07/30/2024] Open
Abstract
Copper (Cu), an essential micronutrient, participates in several physiological processes, including cell proliferation and development. Notably, the disturbance of Cu homeostasis promotes tumor progression through the generation of oxidative stress. Chronic or excessive accumulation of reactive oxygen species (ROS) causes lipid peroxidation, protein denaturation, and enzyme inactivation, which leads to a breakdown of intracellular homeostasis and exacerbates tumor progression. The disruption of the ROS scavenging mechanism also reduces resistance to oxidative stress, leading to further deterioration in a disease state, and maintenance of redox homeostasis is thought to inhibit the onset and progression of various diseases. Superoxide dismutase 3 (SOD3), a Cu-containing secretory antioxidative enzyme, plays a key role in extracellular redox regulation, and the significant reduction in SOD3 facilitates tumor progression. Furthermore, the significant induction of SOD3 participates in tumor metastasis. This review focuses on the role of Cu homeostasis and antioxidative enzymes, including SOD3, in tumor progression, to help clarify the role of redox regulation.
Collapse
Affiliation(s)
- Tetsuro Kamiya
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| |
Collapse
|
35
|
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
|
36
|
Kuchur O, Pogodaeva S, Shcherbakova A, Tsymbal S. Atox1-cyclin D1 loop activity is critical for survival of tumor cells with inactivated TP53. Biosci Rep 2024; 44:BSR20240389. [PMID: 38813981 PMCID: PMC11166628 DOI: 10.1042/bsr20240389] [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: 04/03/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 05/31/2024] Open
Abstract
The search for relevant molecular targets is one of the main tasks of modern tumor chemotherapy. To successfully achieve this, it is necessary to have the most complete understanding of the functioning of a transcriptional apparatus of the cell, particularly related to proliferation. The p53 protein plays an important role in regulating processes such as apoptosis, repair, and cell division, and the loss of its functionality often accompanies various types of tumors and contributes to the development of chemoresistance. Additionally, the proliferative activity of tumor cells is closely related to the metabolism of transition metals. For example, the metallochaperone Atox1 - a copper transporter protein - acts as a transcription activator for cyclin D1, promoting progression through the G1/S phase of the cell cycle. On the other hand, p53 suppresses cyclin D1 at the transcriptional level, thereby these proteins have divergent effects on cell cycle progression. However, the contribution of the interaction between these proteins to cell survival is poorly understood. This work demonstrates that not only exists a positive feedback loop between Atox1 and cyclin D1 but also that the activity of this loop depends on the status of the TP53 gene. Upon inactivation of TP53 in A549 and HepG2 cell lines, the expression of ATOX1 and CCND1 genes is enhanced, and their suppression in these cells leads to pronounced apoptosis. This fundamental observation may be useful in selecting more precise interventions for combined therapy of p53-negative tumors.
Collapse
Affiliation(s)
- Oleg A. Kuchur
- National Research University ITMO, 197101 St. Petersburg, Russia
| | | | | | | |
Collapse
|
37
|
Manchia M, Paribello P, Pinna M, Faa G. The Role of Copper Overload in Modulating Neuropsychiatric Symptoms. Int J Mol Sci 2024; 25:6487. [PMID: 38928192 PMCID: PMC11204094 DOI: 10.3390/ijms25126487] [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: 05/10/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Copper is a transition metal essential for growth and development and indispensable for eukaryotic life. This metal is essential to neuronal function: its deficiency, as well as its overload have been associated with multiple neurodegenerative disorders such as Alzheimer's disease and Wilson's disease and psychiatric conditions such as schizophrenia, bipolar disorder, and major depressive disorders. Copper plays a fundamental role in the development and function of the human Central Nervous System (CNS), being a cofactor of multiple enzymes that play a key role in physiology during development. In this context, we thought it would be timely to summarize data on alterations in the metabolism of copper at the CNS level that might influence the development of neuropsychiatric symptoms. We present a non-systematic review with the study selection based on the authors' judgement to offer the reader a perspective on the most significant elements of neuropsychiatric symptoms in Wilson's disease. We highlight that Wilson's disease is characterized by marked heterogeneity in clinical presentation among patients with the same mutation. This should motivate more research efforts to disentangle the role of environmental factors in modulating the expression of genetic predisposition to this disorder.
Collapse
Affiliation(s)
- Mirko Manchia
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy;
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Pasquale Paribello
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy;
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| | - Martina Pinna
- Forensic Psychiatry Unit, Sardinia Health Agency, 09123 Cagliari, Italy;
| | - Gavino Faa
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy;
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
38
|
Wang Y, Pei P, Yang K, Guo L, Li Y. Copper in colorectal cancer: From copper-related mechanisms to clinical cancer therapies. Clin Transl Med 2024; 14:e1724. [PMID: 38804588 PMCID: PMC11131360 DOI: 10.1002/ctm2.1724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Copper, a trace element and vital cofactor, plays a crucial role in the maintenance of biological functions. Recent evidence has established significant correlations between copper levels, cancer development and metastasis. The strong redox-active properties of copper offer both benefits and disadvantages to cancer cells. The intestinal tract, which is primarily responsible for copper uptake and regulation, may suffer from an imbalance in copper homeostasis. Colorectal cancer (CRC) is the most prevalent primary cancer of the intestinal tract and is an aggressive malignant disease with limited therapeutic options. Current research is primarily focused on the relationship between copper and CRC. Innovative concepts, such as cuproplasia and cuproptosis, are being explored to understand copper-related cellular proliferation and death. Cuproplasia is the regulation of cell proliferation that is mediated by both enzymatic and nonenzymatic copper-modulated activities. Whereas, cuproptosis refers to cell death induced by excess copper via promoting the abnormal oligomerisation of lipoylated proteins within the tricarboxylic acid cycle, as well as by diminishing the levels of iron-sulphur cluster proteins. A comprehensive understanding of copper-related cellular proliferation and death mechanisms offers new avenues for CRC treatment. In this review, we summarise the evolving molecular mechanisms, ranging from abnormal intracellular copper concentrations to the copper-related proteins that are being discovered, and discuss the role of copper in the pathogenesis, progression and potential therapies for CRC. Understanding the relationship between copper and CRC will help provide a comprehensive theoretical foundation for innovative treatment strategies in CRC management.
Collapse
Affiliation(s)
- Yuhong Wang
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of PathologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Pei Pei
- State Key Laboratory of Radiation Medicine and ProtectionSchool of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD‐X)Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouJiangsuChina
| | - Kai Yang
- Department of PathologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionSchool of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD‐X)Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouJiangsuChina
| | - Lingchuan Guo
- Department of PathologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yuan Li
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
39
|
Gomes KS, Coelho JA, Gomes RN, Bosquetti LM, Lange CN, Batista BL, Cerchiaro G, Lago JHG. Dehydrodieugenol isolated from Ocotea cymbarum induces cell death in human breast cancer cell lines by dysregulation of intracellular copper concentration. Chem Biol Interact 2024; 396:111039. [PMID: 38719171 DOI: 10.1016/j.cbi.2024.111039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/16/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
In this work, two neolignans - dehydrodieugenol (1) and dehydrodieugenol B (2) - were isolated from leaves of Ocotea cymbarum (H. B. K.) Ness. (Lauraceae). When tested against two human breast cancer cell lines (MCF7 and MDA-MB-231), compound 1 was inactive (IC50 > 500 μM) whereas compound 2 displayed IC50 values of 169 and 174 μM, respectively. To evaluate, for the first time in the literature, the synergic cytotoxic effects of compounds 1 and 2 with ion Cu2+, both cell lines were incubated with equimolar solutions of these neolignans and Cu(ClO4)2·6H2O. Obtained results revealed no differences in cytotoxicity upon the co-administration of compound 2 and Cu2+. However, the combination of compound 1 and Cu2+ increases the cytotoxicity against MCF7 and MDA-MB-231 cells, with IC50 values of 165 and 204 μM, respectively. The activity of compound 1 and Cu2+ in MCF7 spheroids regarding the causes/effects considering the tumoral microenvironment were accessed using fluorescence staining and imaging by fluorescence microscopy. This analysis enabled the observation of a higher red filter fluorescence intensity in the quiescence zone and the necrotic core, indicating a greater presence of dead cells, suggesting that the combination permeates the spheroid. Finally, using ICP-MS analysis, the intracellular copper disbalance caused by mixing compound 1 and Cu2+ was determined quantitatively. The findings showcased a 50-fold surge in the concentration of Cu2+ compared with untreated cells (p > 0.0001) - 18.7 ng of Cu2+/mg of proteins and 0.37 ng of Cu2+/mg of protein, respectively. Conversely, the concentration of Cu2+ in cells treated with compound 1 was similar to values of the negative control group (0.29 ng of Cu2+/mg of protein). This alteration allowed us to infer that compound 1 combined with Cu2+ induces cell death through copper homeostasis dysregulation.
Collapse
Affiliation(s)
- Kaio S Gomes
- Laboratory of Chemical Biology, Centre of Natural Sciences and Humanities, Federal University of ABC, 09280-580, Santo Andre, SP, Brazil; Metal Biochemistry and Oxidative Stress Laboratory, Centre of Natural Sciences and Humanities, Federal University of ABC, 09280-580, Santo Andre, SP, Brazil
| | - Julia A Coelho
- Metal Biochemistry and Oxidative Stress Laboratory, Centre of Natural Sciences and Humanities, Federal University of ABC, 09280-580, Santo Andre, SP, Brazil
| | - Rafael N Gomes
- Metal Biochemistry and Oxidative Stress Laboratory, Centre of Natural Sciences and Humanities, Federal University of ABC, 09280-580, Santo Andre, SP, Brazil
| | - Lucas M Bosquetti
- Metal Biochemistry and Oxidative Stress Laboratory, Centre of Natural Sciences and Humanities, Federal University of ABC, 09280-580, Santo Andre, SP, Brazil
| | - Camila N Lange
- Environmentals BR, Centre of Natural Sciences and Humanities, Federal University of ABC, 09280-580, Santo Andre, SP, Brazil
| | - Bruno L Batista
- Environmentals BR, Centre of Natural Sciences and Humanities, Federal University of ABC, 09280-580, Santo Andre, SP, Brazil
| | - Giselle Cerchiaro
- Metal Biochemistry and Oxidative Stress Laboratory, Centre of Natural Sciences and Humanities, Federal University of ABC, 09280-580, Santo Andre, SP, Brazil.
| | - João Henrique G Lago
- Laboratory of Chemical Biology, Centre of Natural Sciences and Humanities, Federal University of ABC, 09280-580, Santo Andre, SP, Brazil.
| |
Collapse
|
40
|
Chen M, Chen Y, Fu R, Liu S, Li H, Shen T. Atox1 regulates macrophage polarization in intestinal inflammation via ROS-NLRP3 inflammasome pathway. J Transl Med 2024; 22:497. [PMID: 38796413 PMCID: PMC11128112 DOI: 10.1186/s12967-024-05314-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
BACKGROUND Inflammation and oxidative stress play an important role in the pathophysiology of inflammatory bowel disease (IBD). This study aimed to explore the effects of copper chaperone Antioxidant-1 (Atox1) on macrophages in a mouse model of intestinal inflammation. METHODS A mouse model of TNBS-induced colitis was established and verified using the disease activity index. Atox1 conditional knockout mice were applied. The proportion of macrophages in colonic lamina propria mononuclear cells and ROS production were analyzed using flow cytometry. Inflammatory cytokines were measured using ELISA. Expression of macrophage M1/M2 polarization markers, p47phox, NLRP3, and Caspase-1 p20 was measured using quantitative RT-PCR and Western blotting. RESULTS Atox1 expression was up-regulated in colon tissues of TNBS-induced colitis mice. Macrophages isolated from TNBS-induced colitis mice showed M1 polarization and nuclear translocation of Atox1. Inhibiting copper chaperone activity decreased p47phox, ROS production, and M1 polarization induced by CuCl2 in macrophages. TNBS induced up-regulation of inflammatory cytokines, M1 polarization markers, and p47phox expression in mice, an effect which was preempted by Atox1 knockout. Inflammatory cytokines and expression of M1 polarization markers, p47phox, NLRP3, Caspase-1 p20 were also increased in macrophages isolated from TNBS-induced colitis mice. These changes were alleviated in mice with Atox1 knockout. The effects of Atox1 on macrophage polarization were mediated via the ROS-NLRP3 inflammasome pathway. CONCLUSION Atox1 plays a pro-inflammatory role, promotes M1 polarization of macrophages, and increases the concentrations of pro-inflammatory cytokines in intestinal tissue by regulating the ROS-NLRP3 inflammasome pathway. Atox1 is a potential therapeutic target in IBD.
Collapse
Affiliation(s)
- MingXian Chen
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, No. 234, Gucui road, Hangzhou, 310012, China
- Institute of Integrated Chinese and Western Medicine on Spleen-Stomach Diseases, Zhejiang Province Academy of Traditional Chinese Medicine, Hangzhou, 310012, China
| | - Yu Chen
- Laboratory Animal Center, Zhejiang Province Academy of Traditional Chinese Medicine, Hangzhou, 310012, China
| | - Rui Fu
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, No. 234, Gucui road, Hangzhou, 310012, China
| | - SaiYue Liu
- Department of Adverse Drug Reaction Monitoring, Zhejiang Province Center of Adverse Drug Reaction Monitoring, No. 39, Yile road, Hangzhou, 310012, China.
| | - HaiXia Li
- Department of Cardiology, Guanganmen Hospital of China Academy of Chinese Medical Sciences, No. 5, Beixian Ge, Xicheng District, Beijing, 100053, China.
| | - TangBiao Shen
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, No. 234, Gucui road, Hangzhou, 310012, China.
| |
Collapse
|
41
|
Verdejo-Torres O, Klein DC, Novoa-Aponte L, Carrazco-Carrillo J, Bonilla-Pinto D, Rivera A, Fitisemanu F, Jiménez-González ML, Flinn L, Pezacki AT, Lanzirotti A, Ortiz-Frade LA, Chang CJ, Navea JG, Blaby-Haas C, Hainer SJ, Padilla-Benavides T. Cysteine Rich Intestinal Protein 2 is a copper-responsive regulator of skeletal muscle differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592485. [PMID: 38746126 PMCID: PMC11092763 DOI: 10.1101/2024.05.03.592485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Copper (Cu) is an essential trace element required for respiration, neurotransmitter synthesis, oxidative stress response, and transcriptional regulation. Imbalance in Cu homeostasis can lead to several pathological conditions, affecting neuronal, cognitive, and muscular development. Mechanistically, Cu and Cu-binding proteins (Cu-BPs) have an important but underappreciated role in transcription regulation in mammalian cells. In this context, our lab investigates the contributions of novel Cu-BPs in skeletal muscle differentiation using murine primary myoblasts. Through an unbiased synchrotron X-ray fluorescence-mass spectrometry (XRF/MS) metalloproteomic approach, we identified the murine cysteine rich intestinal protein 2 (mCrip2) in a sample that showed enriched Cu signal, which was isolated from differentiating primary myoblasts derived from mouse satellite cells. Immunolocalization analyses showed that mCrip2 is abundant in both nuclear and cytosolic fractions. Thus, we hypothesized that mCrip2 might have differential roles depending on its cellular localization in the skeletal muscle lineage. mCrip2 is a LIM-family protein with 4 conserved Zn2+-binding sites. Homology and phylogenetic analyses showed that mammalian Crip2 possesses histidine residues near two of the Zn2+-binding sites (CX2C-HX2C) which are potentially implicated in Cu+-binding and competition with Zn2+. Biochemical characterization of recombinant human hsCRIP2 revealed a high Cu+-binding affinity for two and four Cu+ ions and limited redox potential. Functional characterization using CRISPR/Cas9-mediated deletion of mCrip2 in primary myoblasts did not impact proliferation, but impaired myogenesis by decreasing the expression of differentiation markers, possibly attributed to Cu accumulation. Transcriptome analyses of proliferating and differentiating mCrip2 KO myoblasts showed alterations in mRNA processing, protein translation, ribosome synthesis, and chromatin organization. CUT&RUN analyses showed that mCrip2 associates with a select set of gene promoters, including MyoD1 and metallothioneins, acting as a novel Cu-responsive or Cu-regulating protein. Our work demonstrates novel regulatory functions of mCrip2 that mediate skeletal muscle differentiation, presenting new features of the Cu-network in myoblasts.
Collapse
Affiliation(s)
- Odette Verdejo-Torres
- Department of Molecular Biology and Biochemistry, Wesleyan University, CT, 06459. USA
| | - David C. Klein
- Department of Biological Sciences. University of Pittsburgh, Pittsburgh, PA. 15207. USA
| | - Lorena Novoa-Aponte
- Present address: Genetics and Metabolism Section, Liver Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD. USA
| | | | - Denzel Bonilla-Pinto
- Department of Molecular Biology and Biochemistry, Wesleyan University, CT, 06459. USA
| | - Antonio Rivera
- Department of Molecular Biology and Biochemistry, Wesleyan University, CT, 06459. USA
| | | | | | - Lyra Flinn
- Chemistry Department. Skidmore College, Saratoga Springs New York, 12866. USA
| | - Aidan T. Pezacki
- Department of Chemistry. University of California, Berkeley, California, 94720. USA
| | - Antonio Lanzirotti
- Center for Advanced Radiation Sources, The University of Chicago, Lemont, IL 60439. USA
| | | | - Christopher J. Chang
- Department of Chemistry. University of California, Berkeley, California, 94720. USA
- Department of Molecular and Cell Biology. University of California, Berkeley, California, 94720. USA
| | - Juan G. Navea
- Chemistry Department. Skidmore College, Saratoga Springs New York, 12866. USA
| | - Crysten Blaby-Haas
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA & DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA. USA
| | - Sarah J. Hainer
- Department of Biological Sciences. University of Pittsburgh, Pittsburgh, PA. 15207. USA
| | | |
Collapse
|
42
|
Tomasello B, Bellia F, Naletova I, Magrì A, Tabbì G, Attanasio F, Tomasello MF, Cairns WRL, Fortino M, Pietropaolo A, Greco V, La Mendola D, Sciuto S, Arena G, Rizzarelli E. BDNF- and VEGF-Responsive Stimulus to an NGF Mimic Cyclic Peptide with Copper Ionophore Capability and Ctr1/CCS-Driven Signaling. ACS Chem Neurosci 2024; 15:1755-1769. [PMID: 38602894 DOI: 10.1021/acschemneuro.3c00716] [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] [Indexed: 04/13/2024] Open
Abstract
Neurotrophins are a family of growth factors that play a key role in the development and regulation of the functioning of the central nervous system. Their use as drugs is made difficult by their poor stability, cellular permeability, and side effects. Continuing our effort to use peptides that mimic the neurotrophic growth factor (NGF), the family model protein, and specifically the N-terminus of the protein, here we report on the spectroscopic characterization and resistance to hydrolysis of the 14-membered cyclic peptide reproducing the N-terminus sequence (SSSHPIFHRGEFSV (c-NGF(1-14)). Far-UV CD spectra and a computational study show that this peptide has a rigid conformation and left-handed chirality typical of polyproline II that favors its interaction with the D5 domain of the NGF receptor TrkA. c-NGF(1-14) is able to bind Cu2+ with good affinity; the resulting complexes have been characterized by potentiometric and spectroscopic measurements. Experiments on PC12 cells show that c-NGF(1-14) acts as an ionophore, influencing the degree and the localization of both the membrane transporter (Ctr1) and the copper intracellular transporter (CCS). c-NGF(1-14) induces PC12 differentiation, mimics the protein in TrkA phosphorylation, and activates the kinase cascade, inducing Erk1/2 phosphorylation. c-NGF(1-14) biological activities are enhanced when the peptide interacts with Cu2+ even with the submicromolar quantities present in the culture media as demonstrated by ICP-OES measurements. Finally, c-NGF(1-14) and Cu2+ concur to activate the cAMP response element-binding protein CREB that, in turn, induces the brain-derived neurotrophic factor (BDNF) and the vascular endothelial growth factor (VEGF) release.
Collapse
Affiliation(s)
- Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, V.le Andrea Doria 6, Catania 95125, Italy
| | - Francesco Bellia
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | - Irina Naletova
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | - Antonio Magrì
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | - Giovanni Tabbì
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | | | | | - Warren R L Cairns
- Istituto di Scienze Polari (ISP), c/o Campus Scientifico, Università Ca' Foscari Venezia Via Torino, Venezia Mestre 155-30170, Italy
| | - Mariagrazia Fortino
- Dipartimento di Scienze della Salute, Università di Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute, Università di Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| | - Diego La Mendola
- Department of Pharmaceutical Sciences, University of Pisa, Bonanno Pisano 12, Pisa 56126, Italy
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| | - Giuseppe Arena
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| | - Enrico Rizzarelli
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| |
Collapse
|
43
|
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: 96] [Impact Index Per Article: 96.0] [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
|
44
|
Ban XX, Wan H, Wan XX, Tan YT, Hu XM, Ban HX, Chen XY, Huang K, Zhang Q, Xiong K. Copper Metabolism and Cuproptosis: Molecular Mechanisms and Therapeutic Perspectives in Neurodegenerative Diseases. Curr Med Sci 2024; 44:28-50. [PMID: 38336987 DOI: 10.1007/s11596-024-2832-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/17/2023] [Indexed: 02/12/2024]
Abstract
Copper is an essential trace element, and plays a vital role in numerous physiological processes within the human body. During normal metabolism, the human body maintains copper homeostasis. Copper deficiency or excess can adversely affect cellular function. Therefore, copper homeostasis is stringently regulated. Recent studies suggest that copper can trigger a specific form of cell death, namely, cuproptosis, which is triggered by excessive levels of intracellular copper. Cuproptosis induces the aggregation of mitochondrial lipoylated proteins, and the loss of iron-sulfur cluster proteins. In neurodegenerative diseases, the pathogenesis and progression of neurological disorders are linked to copper homeostasis. This review summarizes the advances in copper homeostasis and cuproptosis in the nervous system and neurodegenerative diseases. This offers research perspectives that provide new insights into the targeted treatment of neurodegenerative diseases based on cuproptosis.
Collapse
Affiliation(s)
- Xiao-Xia Ban
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 430013, China
| | - Hao Wan
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 430013, China
| | - Xin-Xing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha, 430013, China
| | - Ya-Ting Tan
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 430013, China
| | - Xi-Min Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 430013, China
| | - Hong-Xia Ban
- Affiliated Hospital, Inner Mongolia Medical University, Hohhot, 010050, China
| | - Xin-Yu Chen
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 430013, China
| | - Kun Huang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 430013, China
| | - Qi Zhang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 430013, China.
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China.
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 430013, China.
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China.
- Hunan Key Laboratory of Ophthalmology, Changsha, 430013, China.
| |
Collapse
|
45
|
Pan Q, Chen C, Yang YJ. Top Five Stories of the Cellular Landscape and Therapies of Atherosclerosis: Current Knowledge and Future Perspectives. Curr Med Sci 2024; 44:1-27. [PMID: 38057537 DOI: 10.1007/s11596-023-2818-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/22/2023] [Indexed: 12/08/2023]
Abstract
Atherosclerosis (AS) is characterized by impairment and apoptosis of endothelial cells, continuous systemic and focal inflammation and dysfunction of vascular smooth muscle cells, which is documented as the traditional cellular paradigm. However, the mechanisms appear much more complicated than we thought since a bulk of studies on efferocytosis, transdifferentiation and novel cell death forms such as ferroptosis, pyroptosis, and extracellular trap were reported. Discovery of novel pathological cellular landscapes provides a large number of therapeutic targets. On the other side, the unsatisfactory therapeutic effects of current treatment with lipid-lowering drugs as the cornerstone also restricts the efforts to reduce global AS burden. Stem cell- or nanoparticle-based strategies spurred a lot of attention due to the attractive therapeutic effects and minimized adverse effects. Given the complexity of pathological changes of AS, attempts to develop an almighty medicine based on single mechanisms could be theoretically challenging. In this review, the top stories in the cellular landscapes during the initiation and progression of AS and the therapies were summarized in an integrated perspective to facilitate efforts to develop a multi-targets strategy and fill the gap between mechanism research and clinical translation. The future challenges and improvements were also discussed.
Collapse
Affiliation(s)
- Qi Pan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Cheng Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
46
|
Yang S, Li Y, Zhou L, Wang X, Liu L, Wu M. Copper homeostasis and cuproptosis in atherosclerosis: metabolism, mechanisms and potential therapeutic strategies. Cell Death Discov 2024; 10:25. [PMID: 38218941 PMCID: PMC10787750 DOI: 10.1038/s41420-023-01796-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 01/15/2024] Open
Abstract
Copper is an essential micronutrient that plays a pivotal role in numerous physiological processes in virtually all cell types. Nevertheless, the dysregulation of copper homeostasis, whether towards excess or deficiency, can lead to pathological alterations, such as atherosclerosis. With the advent of the concept of copper-induced cell death, termed cuproptosis, researchers have increasingly focused on the potential role of copper dyshomeostasis in atherosclerosis. In this review, we provide a broad overview of cellular and systemic copper metabolism. We then summarize the evidence linking copper dyshomeostasis to atherosclerosis and elucidate the potential mechanisms underlying atherosclerosis development in terms of both copper excess and copper deficiency. Furthermore, we discuss the evidence for and mechanisms of cuproptosis, discuss its interactions with other modes of cell death, and highlight the role of cuproptosis-related mitochondrial dysfunction in atherosclerosis. Finally, we explore the therapeutic strategy of targeting this novel form of cell death, aiming to provide some insights for the management of atherosclerosis.
Collapse
Affiliation(s)
- Shengjie Yang
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yujuan Li
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Lijun Zhou
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xinyue Wang
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Longtao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Min Wu
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
47
|
Wang Y, Chen Y, Zhang J, Yang Y, Fleishman JS, Wang Y, Wang J, Chen J, Li Y, Wang H. Cuproptosis: A novel therapeutic target for overcoming cancer drug resistance. Drug Resist Updat 2024; 72:101018. [PMID: 37979442 DOI: 10.1016/j.drup.2023.101018] [Citation(s) in RCA: 85] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/20/2023]
Abstract
Cuproptosis is a newly identified form of cell death driven by copper. Recently, the role of copper and copper triggered cell death in the pathogenesis of cancers have attracted attentions. Cuproptosis has garnered enormous interest in cancer research communities because of its great potential for cancer therapy. Copper-based treatment exerts an inhibiting role in tumor growth and may open the door for the treatment of chemotherapy-insensitive tumors. In this review, we provide a critical analysis on copper homeostasis and the role of copper dysregulation in the development and progression of cancers. Then the core molecular mechanisms of cuproptosis and its role in cancer is discussed, followed by summarizing the current understanding of copper-based agents (copper chelators, copper ionophores, and copper complexes-based dynamic therapy) for cancer treatment. Additionally, we summarize the emerging data on copper complexes-based agents and copper ionophores to subdue tumor chemotherapy resistance in different types of cancers. We also review the small-molecule compounds and nanoparticles (NPs) that may kill cancer cells by inducing cuproptosis, which will shed new light on the development of anticancer drugs through inducing cuproptosis in the future. Finally, the important concepts and pressing questions of cuproptosis in future research that should be focused on were discussed. This review article suggests that targeting cuproptosis could be a novel antitumor therapy and treatment strategy to overcome cancer drug resistance.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, PR China.
| | - Yongming Chen
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Junjing Zhang
- Department of Hepato-Biliary Surgery, Department of Surgery, Huhhot First Hospital, Huhhot 010030, PR China
| | - Yihui Yang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yan Wang
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, PR China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, PR China
| | - Yuanfang Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, PR China.
| | - Hongquan Wang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| |
Collapse
|
48
|
Chen Z, Li YY, Liu X. Copper homeostasis and copper-induced cell death: Novel targeting for intervention in the pathogenesis of vascular aging. Biomed Pharmacother 2023; 169:115839. [PMID: 37976889 DOI: 10.1016/j.biopha.2023.115839] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/25/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
Copper-induced cell death, also known as cuproptosis, is distinct from other types of cell death such as apoptosis, necrosis, and ferroptosis. It can trigger the accumulation of lethal reactive oxygen species, leading to the onset and progression of aging. The significant increases in copper ion levels in the aging populations confirm a close relationship between copper homeostasis and vascular aging. On the other hand, vascular aging is also closely related to the occurrence of various cardiovascular diseases throughout the aging process. However, the specific causes of vascular aging are not clear, and different living environments and stress patterns can lead to individualized vascular aging. By exploring the correlations between copper-induced cell death and vascular aging, we can gain a novel perspective on the pathogenesis of vascular aging and enhance the prognosis of atherosclerosis. This article aims to provide a comprehensive review of the impacts of copper homeostasis on vascular aging, including their effects on endothelial cells, smooth muscle cells, oxidative stress, ferroptosis, intestinal flora, and other related factors. Furthermore, we intend to discuss potential strategies involving cuproptosis and provide new insights for copper-related vascular aging.
Collapse
Affiliation(s)
- Zhuoying Chen
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Yuan-Yuan Li
- Department of Nursing, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China.
| | - Xiangjie Liu
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China.
| |
Collapse
|
49
|
Tsymbal S, Refeld A, Zatsepin V, Kuchur O. The p53 protein is a suppressor of Atox1 copper chaperon in tumor cells under genotoxic effects. PLoS One 2023; 18:e0295944. [PMID: 38127999 PMCID: PMC10735018 DOI: 10.1371/journal.pone.0295944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023] Open
Abstract
The p53 protein is crucial for regulating cell survival and apoptosis in response to DNA damage. However, its influence on therapy effectiveness is controversial: when DNA damage is high p53 directs cells toward apoptosis, while under moderate genotoxic stress it saves the cells from death and promote DNA repair. Furthermore, these processes are influenced by the metabolism of transition metals, particularly copper since they serve as cofactors for critical enzymes. The metallochaperone Atox1 is under intensive study in this context because it serves as transcription factor allegedly mediating described effects of copper. Investigating the interaction between p53 and Atox1 could provide insights into tumor cell survival and potential therapeutic applications in oncology. This study explores the relationship between p53 and Atox1 in HCT116 and A549 cell lines with wild type and knockout TP53. The study found an inverse correlation between Atox1 and p53 at the transcriptional and translational levels in response to genotoxic stress. Atox1 expression decreased with increased p53 activity, while cells with inactive p53 had significantly higher levels of Atox1. Suppression of both genes increased apoptosis, while suppression of the ATOX1 gene prevented apoptosis even under the treatment with chemotherapeutic drugs. The findings suggest that Atox1 may act as one of key elements in promotion of cell cycle under DNA-damaging conditions, while p53 works as an antagonist by inhibiting Atox1. Understanding of this relationship could help identify potential targets in cell signaling pathways to enhance the effectiveness of combined antitumor therapy, especially in tumors with mutant or inactive p53.
Collapse
Affiliation(s)
- Sergey Tsymbal
- International Institute ‘Solution Chemistry of Advanced Materials and Technologies’, ITMO University, St. Petersburg, Russia
| | - Aleksandr Refeld
- International Institute ‘Solution Chemistry of Advanced Materials and Technologies’, ITMO University, St. Petersburg, Russia
| | | | - Oleg Kuchur
- International Institute ‘Solution Chemistry of Advanced Materials and Technologies’, ITMO University, St. Petersburg, Russia
| |
Collapse
|
50
|
Conforti RA, Delsouc MB, Zorychta E, Telleria CM, Casais M. Copper in Gynecological Diseases. Int J Mol Sci 2023; 24:17578. [PMID: 38139406 PMCID: PMC10743751 DOI: 10.3390/ijms242417578] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Copper (Cu) is an essential micronutrient for the correct development of eukaryotic organisms. This metal plays a key role in many cellular and physiological activities, including enzymatic activity, oxygen transport, and cell signaling. Although the redox activity of Cu is crucial for enzymatic reactions, this property also makes it potentially toxic when found at high levels. Due to this dual action of Cu, highly regulated mechanisms are necessary to prevent both the deficiency and the accumulation of this metal since its dyshomeostasis may favor the development of multiple diseases, such as Menkes' and Wilson's diseases, neurodegenerative diseases, diabetes mellitus, and cancer. As the relationship between Cu and cancer has been the most studied, we analyze how this metal can affect three fundamental processes for tumor progression: cell proliferation, angiogenesis, and metastasis. Gynecological diseases are characterized by high prevalence, morbidity, and mortality, depending on the case, and mainly include benign and malignant tumors. The cellular processes that promote their progression are affected by Cu, and the mechanisms that occur may be similar. We analyze the crosstalk between Cu deregulation and gynecological diseases, focusing on therapeutic strategies derived from this metal.
Collapse
Affiliation(s)
- Rocío A. Conforti
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), San Luis CP D5700HHW, Argentina; (R.A.C.); (M.B.D.)
| | - María B. Delsouc
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), San Luis CP D5700HHW, Argentina; (R.A.C.); (M.B.D.)
| | - Edith Zorychta
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC H3A 2B4, Canada;
| | - Carlos M. Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC H3A 2B4, Canada;
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Marilina Casais
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), San Luis CP D5700HHW, Argentina; (R.A.C.); (M.B.D.)
| |
Collapse
|