1
|
Wang X, Xue Y, Chang L, Zhu X, Liu W, Liang T. The Regulation of Trace Metal Elements in Cancer Ferroptosis. Adv Biol (Weinh) 2025:e2400821. [PMID: 40200790 DOI: 10.1002/adbi.202400821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/11/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, as novel type of regulated cell death that has garnered widespread attention over the past decade, has witnessed the continuous discovery of an increasing number of regulatory mechanisms. Trace metal elements play a multifaceted and crucial role in oncology. Interestingly, it has been increasingly evident that these elements, such as copper, are involved in the regulation of iron accumulation, lipid peroxidation and antiferroptotic systems, suggesting the existence of "nonferrous" mechanisms in ferroptosis. In this review, a comprehensive overview of the composition and mechanism of ferroptosis is provided. The interaction between copper metabolism (including cuproptosis) and ferroptosis in cancer, as well as the roles of other trace metal elements (such as zinc, manganese, cobalt, and molybdenum) in ferroptosis are specifically focused. Furthermore, the applications of nanomaterials based on these metals in cancer therapy are also reviewed and potential strategies for co-targeting ferroptosis and cuproptosis are explored. Nevertheless, in light of the intricate and ambiguous nature of these interactions, ongoing research is essential to further elucidate the "nonferrous" mechanisms of ferroptosis, thereby facilitating the development of novel therapeutic targets and approaches for cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuanyuan Xue
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lei Chang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xuena Zhu
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Wenjun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
- Zhejiang University Cancer Center, Hangzhou, 310003, China
| |
Collapse
|
2
|
Ma M, Pan Z, Zhu Z, Ling C, Yuan J, Huo X, Li S, Liu R. Enhanced cancer therapy using modified magnetic α-Fe 2O 3/Fe 3O 4nanorods: Dual role in curcumin delivery and ferroptosis induction. Colloids Surf B Biointerfaces 2025; 252:114689. [PMID: 40228427 DOI: 10.1016/j.colsurfb.2025.114689] [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: 02/27/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/16/2025]
Abstract
Curcumin (CUR) has gained considerable attention in oncology due to its potent anti-tumor, anti-inflammatory, and antioxidant properties. However, its clinical utility was significantly hindered by low bioavailability and rapid metabolic degradation. This work designed a magnetic α-Fe2O3/Fe3O4 heterogeneous nanorod prepared via a urea hydrolysis-calcination process for curcumin delivery. The nanorods were modified with hyaluronic acid (HA), providing a stable matrix for curcumin encapsulation. The zeta potential of α-Fe2O3/Fe3O4/HA/CUR was -3.94 mV, the saturation magnetization was 7.82 emu·g-1, the encapsulation rate and drug loading rate were 10.53 % and 29.64 % respectively. At pH 5.4, 6.5, and 7.4, the release rates were 44.8 %, 40.2 %, and 39.3 %, respectively. Kinetic modeling indicated that release profiles followed the Weibull kinetic model, with an R2 value greater than 0.98. The α-Fe2O3/Fe3O4/HA/CUR exhibited excellent magnetic responsiveness, demonstrating a significant inhibitory effect on human liver cancer (HepG2) cells (inhibition rate greater than 60 %) under a magnetic field. Moreover, they substantially reduced the cytotoxicity of curcumin on normal human (LO2) cells. The α-Fe2O3/Fe3O4/HA/CUR inhibited migration and proliferation of HepG2 cells, induced apoptosis via the Caspase pathway, and synergistically suppressed tumor cell development through ferroptosis. The drug release from the α-Fe2O3/Fe3O4/HA/CUR was stable, significantly enhancing the bioavailability of curcumin. This provided a promising strategy for improving the bioavailability of poorly soluble drugs and enhancing liver cancer treatment outcomes.
Collapse
Affiliation(s)
- Mingyi Ma
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Zhongjun Pan
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang 212300, PR China
| | - Ziye Zhu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Chen Ling
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Jiahao Yuan
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Xiangdong Huo
- School of Materials Science & Engineering, Jiangsu University, Zhenjiang 212013, PR China.
| | - Shasha Li
- Affiliated Kunshan Hospital, Jiangsu University, Suzhou 215300, PR China.
| | - Ruijiang Liu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
3
|
Wang L, ChenLiu Z, Wang D, Tang D. Cross-talks of GSH, mitochondria, RNA m6A modification, NRF2, and p53 between ferroptosis and cuproptosis in HCC: A review. Int J Biol Macromol 2025; 302:140523. [PMID: 39894098 DOI: 10.1016/j.ijbiomac.2025.140523] [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: 11/11/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with high morbidity and mortality, as well as poor prognosis. Therefore, it is imperative to explore alternative therapeutic targets for HCC treatment. Ferroptosis and cuproptosis have recently been identified as metal-dependent cell death mechanisms that play significant roles in HCC treatment. This study identified potential cross-talk between ferroptosis and cuproptosis, including the common hub glutathione, common site of occurrence, mitochondria, shared epigenetic modification mode, RNA N6 methyladenosine modification, mutual inhibitor, nuclear factor erythroid 2-related factor 2, and dual regulator, p53. These findings provide a theoretical foundation for the joint induction of HCC cell death and effective inhibition of HCC progression. However, some immune cells are susceptible to ferroptosis or cuproptosis, which may impair or enhance anti-cancer immune function. We propose strategies to target specific targets molecules such as tripartite motif containing 25, ferroptosis suppressor protein 1, and peroxisome proliferator-activated receptor gamma or exploit the unique acidic environment surrounding cancer cells to precisely induce ferroptosis in cancer cells. This approach aims to advance the development of precision medicine for HCC treatment.
Collapse
Affiliation(s)
- Leihan Wang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Zhenni ChenLiu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China.
| |
Collapse
|
4
|
Yang L, Xie L, Li M, Miao Y, Yang J, Chen S, Ma X, Xie P. Potential relationship between cuproptosis and sepsis-acquired weakness: an intermediate role for mitochondria. Front Physiol 2025; 16:1520669. [PMID: 40182687 PMCID: PMC11965645 DOI: 10.3389/fphys.2025.1520669] [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: 10/31/2024] [Accepted: 03/05/2025] [Indexed: 04/05/2025] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. Skeletal muscle atrophy due to critical illness is a common phenomenon in the intensive care unit (ICU) and is referred to as ICU-acquired weakness (ICU-AW). The occurrence of ICU-AW in patients with sepsis is known as sepsis-acquired weakness (SAW). Furthermore, it is well known that maintaining normal muscle function closely relates to mitochondrial homeostasis. Once mitochondrial function is impaired, both muscle quality and function are affected. Copper plays a key role in mitochondrial homeostasis as a transition metal that regulates the function and stability of various enzymes. Copper is also involved in oxidation-reduction reactions, and intracellular copper overload causes oxidative stress and induces cell death. Previous studies have shown that excess intracellular copper induces cell death by targeting lipid-acylated proteins that regulate the mitochondrial tricarboxylic acid (TCA) cycle, which differs from the known canonical mechanisms of regulated cell death. Furthermore, inhibitors of cell death, such as apoptosis, necroptosis, pyroptosis and ferroptosis, are not effective in preventing copper-induced cell death. This new form of cell death has been termed "Cuproptosis"; however, the mechanism by which copper-induced cell death is involved in SAW remains unclear. In this paper, we review the possible relationship between cuproptosis and SAW. Cuproptosis may be involved in regulating the pathological mechanisms of SAW through mitochondria-related signaling pathways, mitochondria-related ferroptosis mechanisms, and mitochondria-related genes, and to provide new ideas for further investigations into the mechanism of SAW.
Collapse
Affiliation(s)
- Luying Yang
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People’s Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| | - Leiyu Xie
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People’s Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| | - Min Li
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People’s Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| | - Yanmei Miao
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People’s Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| | - Jun Yang
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People’s Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| | - Shaolin Chen
- Department of Nursing of Affiliated Hospital, Zunyi Medical University, Zunyi, China
| | - Xinglong Ma
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People’s Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| | - Peng Xie
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People’s Hospital of Zunyi), Zunyi Medical University, Zunyi, China
- Department of Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
5
|
Wei H, Peng J. Integrated Analysis of Bulk and Single-Cell RNA Sequencing Data Reveal a Novel Prognostic Signature of Combining Cuproptosis- and Ferroptosis-Related Genes in Hepatocellular Carcinoma. Int J Mol Sci 2025; 26:2779. [PMID: 40141422 PMCID: PMC11943219 DOI: 10.3390/ijms26062779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
As a common malignancy, hepatocellular carcinoma (HCC) proliferation and metastasis could be promoted by ferroptosis and cuproptosis. In this study, we screened out the differentially expressed cuproptosis- and ferroptosis-related genes (CFRGs) and identified the 17 informative prognosis-associated genes. A CFRG scoring model was constructed based on the subtypes identified by consensus clustering analysis and principal component analysis (PCA). Furthermore, the immune profile, expression of immune checkpoint genes (ICGs) and drug susceptibility were also compared between the two CFRG score groups. The results showed that patients with a high CFRG score had higher survival probabilities. The correlation analysis suggested that CFRG scores were negatively correlated with activated CD4.T.cell. The expression patterns of thirty ICGs and the half-maximal inhibitory concentration (IC50) values of 128 drugs displayed significant differences between the two CFRG score groups. A statistically significant difference in the efficacy of sorafenib was found between the two CFRG score groups. Moreover, based on multivariate COX regression analysis and weighted gene co-expression network analysis (WGCNA), we screened DLAT and SLC2A1 as signature genes. Molecular docking analysis revealed that DLAT and SLC2A1 had a strong binding affinity toward camptothecin, rapamycin, dactolisib, and luminespib. The correlation between the CFRG score and single-cell characteristics was further explored. The study depended on our understanding of the biological function of CFRGs in HCC and provided new insights for developing treatment strategies.
Collapse
Affiliation(s)
- Hua Wei
- School of Resources and Environmental Science and Engineering, Hubei University of Science and Technology, Xianning 437100, China
- Research Center of Beidou, Industrial Development of Key Research Institute of Humanities and Social Sciences of Hubei Province, Hubei University of Science and Technology, Xianning 437100, China
| | - Jiaxin Peng
- School of Computer Science, National Unversity of Defense Technology, Changsha 410073, China;
| |
Collapse
|
6
|
Gu L, Sun Y, Bai T, Shao S, Tang S, Xue P, Cai W, Qin X, Zeng X, Yan S. Functional nanozyme system for synergistic tumor immunotherapy via cuproptosis and ferroptosis activation. J Nanobiotechnology 2025; 23:212. [PMID: 40089774 PMCID: PMC11909888 DOI: 10.1186/s12951-025-03284-3] [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: 12/14/2024] [Accepted: 03/02/2025] [Indexed: 03/17/2025] Open
Abstract
Elevated copper levels induce tumor cuproptosis and ferroptosis, leading to immunogenic cell death and subsequent antitumor immune responses. However, dysregulated copper metabolism in tumor cells maintains homeostatic copper balance, while hypoxic microenvironments hinder therapeutic efficacy. In this study, we present a nanozyme system, termed CussOMEp, comprising a copper-based nanovector (CussNV) that is PEGylated and loaded with omeprazole, a copper transporter inhibitor, to enhance tumor synergistic immunotherapy by promoting cuproptosis and ferroptosis. CussNV is assembled from dithiodiglycolic acid and copper ions, exhibiting peroxidase, glutathione oxidase, and catalase-like activities, along with responsive degradability. This nanozyme alleviates tumor hypoxia by producing oxygen, induces ferroptosis through the generation of lethal hydroxyl radicals, and depletes glutathione. Additionally, omeprazole increases cellular copper concentration and oxidative stress by inhibiting the intracellular copper-transporting ATPase 1 (ATP7A), enhancing lipoylated protein oligomerization and cuproptosis. In a breast tumor mouse model, CussOMEp elicits robust antitumor immune responses, including dendritic cell maturation and T cell proliferation. When combined with PD-1 antibodies (αPD-1), CussOMEp significantly inhibits tumor metastasis in bilateral and lung metastatic models. This work presents a functional nanozyme system as a promising strategy for synergistic tumor immunotherapy leveraging ferroptosis and cuproptosis.
Collapse
Affiliation(s)
- Lina Gu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150076, China
| | - Ying Sun
- Fujian Key Laboratory of Flexible Electronics, Strait Laboratory of Flexible Electronics (SLoFE), Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
- Department of Gastroenterology, No. 1 Hospital Affiliated with Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Tingjie Bai
- Fujian Key Laboratory of Flexible Electronics, Strait Laboratory of Flexible Electronics (SLoFE), Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Sijie Shao
- Fujian Key Laboratory of Flexible Electronics, Strait Laboratory of Flexible Electronics (SLoFE), Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Shumin Tang
- Key Laboratory of Microbial Pathogenesis, Biomedical Research Center of South China, College of Life Sciences, Interventions of Fujian Province University, Fujian Normal University, 1 Keji Road, Fuzhou, Fujian, 350117, China
| | - Panpan Xue
- Fujian Key Laboratory of Flexible Electronics, Strait Laboratory of Flexible Electronics (SLoFE), Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Wanlin Cai
- Fujian Key Laboratory of Flexible Electronics, Strait Laboratory of Flexible Electronics (SLoFE), Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Xian Qin
- Fujian Key Laboratory of Flexible Electronics, Strait Laboratory of Flexible Electronics (SLoFE), Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Xuemei Zeng
- Key Laboratory of Microbial Pathogenesis, Biomedical Research Center of South China, College of Life Sciences, Interventions of Fujian Province University, Fujian Normal University, 1 Keji Road, Fuzhou, Fujian, 350117, China.
| | - Shuangqian Yan
- Fujian Key Laboratory of Flexible Electronics, Strait Laboratory of Flexible Electronics (SLoFE), Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China.
| |
Collapse
|
7
|
Lu X, Chen Y, Mo Y, Zeng Q, Cen S, Zeng L, Hu H, Li A, Gao X, Zhang B. Atomically dispersed copper(I) on tungstosilicic acid for catalytic protection against cisplatin-induced hearing loss. J Mater Chem B 2025; 13:3540-3552. [PMID: 39991827 DOI: 10.1039/d4tb02820a] [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: 02/25/2025]
Abstract
The employment of platinum-based drugs for cancer chemotherapy, which might yield oxidative stress, is regarded as one main factor leading to hearing loss. The exact molecular mechanisms for cisplatin-induced hearing loss require further clarification, thus limiting the development of FDA-approved therapies. Herein, we mimicked the molecular structure of natural antioxidative enzymes to fabricate a four-oxygen-coordinating copper single-atom nanozyme (Cu SAN) exhibiting good superoxide dismutase and catalase activity, to alleviate the oxidative stress induced by platinum-based drugs. Notably, Cu SAN exhibited profound protective effects against cisplatin-induced hair cell damage with only 15 ng mL-1 of Cu species, successfully reversing cisplatin-induced hearing loss via oral administration. Due to its oxidation resistance, pretreatment with Cu SAN significantly improved cell viability and reduced ROS accumulation in cisplatin-triggered hair cell damage in HEI-OC1 cells and cochlear explants. Our results first demonstrated that cisplatin treatment induced cuproptosis in hair cells by modulating copper ion homeostasis. Further investigation revealed that Cu SAN nanozyme effectively alleviated hair cell cuproptosis by regulating FDX1 and reducing aggregated lipoacylated protein. This research underscores the promising potential of four-oxygen-coordinating Cu nanomaterials as a therapeutic approach to combat hearing loss, providing a new strategy for auditory protection.
Collapse
Affiliation(s)
- Xiaochan Lu
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, Shenzhen, 518036, China.
| | - Yin Chen
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing, 210008, China.
- Department of Otolaryngology and Head-and-Neck Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, 210008, China
| | - Yanmei Mo
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, Shenzhen, 518036, China.
| | - Qingdong Zeng
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, Shenzhen, 518036, China.
| | - Shaoqin Cen
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing, 210008, China.
- Department of Otolaryngology and Head-and-Neck Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, 210008, China
| | - Li Zeng
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, Shenzhen, 518036, China.
| | - Hongyi Hu
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, Shenzhen, 518036, China.
| | - Ao Li
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing, 210008, China.
- Department of Otolaryngology and Head-and-Neck Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, 210008, China
| | - Xia Gao
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing, 210008, China.
- Department of Otolaryngology and Head-and-Neck Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, 210008, China
| | - Bin Zhang
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, Shenzhen, 518036, China.
| |
Collapse
|
8
|
Jin G, Han W, Duan T, Xue Z, Song C, Xu Y, Yu M. Edaravone dextranol alleviates ferroptosis, Cuproptosis, and blood-brain barrier damage after acute cerebral infarction. Metab Brain Dis 2025; 40:134. [PMID: 40029474 DOI: 10.1007/s11011-025-01559-0] [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: 04/30/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Edaravone dextrose (EDB) is a commonly used clinical treatment for cerebral infarction due to its anti-inflammatory and free radical scavenging properties. However, its potential additional neuroprotective mechanisms need to be further investigated. In this study, we evaluated the effects of EDB on ferroptosis, cuproptosis, and blood-brain barrier (BBB) disruption after cerebral infarction in vivo and in vitro by constructing a mouse middle cerebral artery occlusion (MCAO) model, and an oxygen-glucose deprivation/reperfusion (OGD/R) model of neurons and brain microvascular endothelium. Our results showed that EDB treatment improved neurological impairment and brain histopathology in MCAO mice. EDB treatment significantly alleviated ferroptosis and cuproptosis in MCAO mice and OGD/R models of neuronal, in vitro and in vivo, the protective pathway of ferroptosis SLC7A11/GPX4 was detected to be activated and the cuproptosis-promoting protein SLC31A1 and FDX1 were down-regulated. We also found that EDB treatment ameliorated BBB damage in MCAO mice. The endothelial cell protective pathway PDGFRβ/PI3K/AKT activation was also detected in MCAO mice and OGD/R models of endothelial cells after EDB treatment. In conclusion, our study demonstrated that EDB has a good ameliorating effect on ferroptosis, cuproptosis, and BBB damage after cerebral infarction, which provides more evidence for the clinical application of EDB and provides new theories and directions for the study of the mechanism of EDB.
Collapse
Affiliation(s)
- Guimin Jin
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Wei Han
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Tingting Duan
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Zongwei Xue
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Chenglin Song
- Nutritional Department, The Second People'S Hospital of Lianyungang, Lianyungang, 222000, China.
| | - Yuhao Xu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
| | - Ming Yu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
| |
Collapse
|
9
|
Liu Y, Li C, Cui X, Liu C, Xiao P, Yang W. Kynureninase induce cuproptosis resistance in gastric cancer progression through downregulating lipotic acid synthetase mediated non-canonical mechanism. Cell Signal 2025; 127:111565. [PMID: 39681223 DOI: 10.1016/j.cellsig.2024.111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND Gastric cancer (GC) is among the most malignant tumors, with the lowest five-year survival rate, and limited treatment options. Kynureninase (KYNU), is a key molecule in tryptophan metabolism and promotes tumor progression and immunosuppression. Cuproptosis is a non-apoptotic cell death mechanism, primarily due to oxidative stress caused by copper ion accumulation, that is related to tumor progression and drug resistance. KYNU can inhibit ferroptosis of tumor cells by alleviating oxidative stress. Here, we explored whether KYNU can regulate the biological behavior of GC and cuproptosis. METHODS Expression, prognostic association, and functional analysis of KYNU in GC and tumor-adjacent tissues were analyzed using data from The Cancer Genome Atlas and clinical specimens. Effects of KYNU on proliferation, invasion, metastasis, and cuproptosis of GC cells were detected by CCK8, clone formation, Transwell, and flow cytometry assays. Elesclomol (ES) combined with CuCl2 were used to induce cuproptosis in GC cells. 3-hydroxyanthranilic acid (3-HA) was used to indicate KYNU function. Key cuproptosis genes were detected by qPCR and WB. The effects of KYNU on GC cell behavior and cuproptosis through lipoic acid synthetase (LIAS) were verified by stable overexpression and knockdown of LIAS. RESULTS KYNU is highly expressed in GC, and high KYNU expression is an independent predictor of poor prognosis in patients with GC. KYNU can promote GC cell proliferation, invasion, metastasis, and cuproptosis resistance. 3-HA had a certain inhibitory effect on the expression of LIAS, but it was not significant. KYNU had no effect on the intracellular 3-HA level. KYNU expression was negatively correlated with that of LIAS, and promoted GC cell proliferation, invasion, metastasis, and cuproptosis resistance by downregulating LIAS. CONCLUSIONS KYNU can promote GC proliferation, invasion, metastasis, and cuproptosis resistance.This effect is not associated with its metabolite 3-HA, but is achieved by a non classical mechanisms that downregulating the expression of LIAS, a key gene of cuproptosis.
Collapse
Affiliation(s)
- Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Xilun Cui
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pengtuo Xiao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
10
|
Liu XH, Wang GR, Zhong NN, Zhu ZR, Xiao Y, Li Z, Bu LL, Liu B. Metal-dependent cell death resistance contribute to lymph node metastasis of oral squamous cell carcinoma. Front Cell Dev Biol 2025; 13:1541582. [PMID: 40083663 PMCID: PMC11903458 DOI: 10.3389/fcell.2025.1541582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Objectives Ferroptosis and cuproptosis can be summarized as metal-dependent cell death. This study aimed to explore the expression of metal-dependent cell death resistance (MCDR) characteristics in tumor cells of oral squamous cell carcinoma (OSCC) and to explore its relationship with lymph node metastasis (LNM). Methods By integrating single-cell data of OSCC from public databases, an expression matrix comprising 127,149 cells was constructed. Gene set scores were calculated using the irGSEA package, and GO and KEGG analyses were performed to identify enriched pathways. The R package monocle3 was employed to calculate the cell trajectory and infer evolutionary patterns. The MuSiC2 package was employed to enable the evaluation of cell proportions. Cell-cell interaction information was analyzed using the CellChat package. The expression of cathepsin V (CTSV), glutathione peroxidase 4 (GPX4), and cyclin-dependent kinase inhibitor 2A (CDKN2A) was validated via immunohistochemistry and multiplex immunohistochemistry in oral mucosa (OM), non-metastatic primary tumors (nPT), and metastatic primary tumors (mPT). Additionally, R package oncoPredict was utilized to identify potential drug sensitivities. Results The malignant cells in OSCC were divided into five subtypes, among which Epi_2 existed more in mPT and had higher MCDR characteristics. In addition, Epi_2 enriched multiple malignant-related pathways such as HEDGEHOG, NOTCH, and MYC. The spatial transcriptome and bulk RNA data verified that the proportion of Epi_2 in mPT was higher than that in nPT. Cell communication analysis showed that the effect of Epi_2 on endothelial cells was enhanced, which was mainly reflected in VEGFR and CXCL signaling pathways. Immunohistochemical results showed that the expression of Epi_2 characteristic markers CTSV and GPX4 in mPT was significantly higher than that in nPT. Multiplex immunohistochemical results showed that the co-expression cells of CTSV, GPX4 and CDKN2A in mPT were more than those in nPT. OSCC patients with high Epi_2 characteristics may have immunotherapy resistance and anti-EGFR treatment resistance. Doramapimod was identified as a sensitive drug. Conclusion There is a type of malignant cells with characteristics of MDCR in OSCC, which is related to LNM and treatment resistance. It provides a predictive marker for early diagnosis of LNM.
Collapse
Affiliation(s)
- Xuan-Hao Liu
- 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, Wuhan University, Wuhan, China
| | - Guang-Rui Wang
- 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, Wuhan University, Wuhan, China
| | - Nian-Nian Zhong
- 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, Wuhan University, Wuhan, China
| | - Zheng-Rui Zhu
- 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, Wuhan University, Wuhan, China
| | - Yao Xiao
- 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, Wuhan University, Wuhan, China
| | - Zheng Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lin-Lin Bu
- 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, Wuhan University, Wuhan, China
- Department of Oral & Maxillofacial - Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- 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, Wuhan University, Wuhan, China
- Department of Oral & Maxillofacial - Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Yang Y, Dong C, Ma X, Wang Y, Li Z, Xu Y, Chen T, Gao C, Ye X, Wu A, Zhang X. Advances in cuproptosis harnessing copper-based nanomaterials for cancer therapy. J Mater Chem B 2025; 13:2978-2999. [PMID: 39901728 DOI: 10.1039/d4tb02746a] [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: 02/05/2025]
Abstract
Cuproptosis, a newly identified programmed cell death form, is characterized by excessive copper accumulation in cells, resulting in mitochondria damage and toxic protein stress, ultimately causing cell death. Given the considerable therapeutic promise of copper toxicity in cancer treatment, copper-based nanomaterials that induce copper death have attracted interest as a promising approach for tumor therapy. This review comprehensively introduces the mechanisms of cuproptosis and the associated regulatory genes, including both positive and negative regulatory regulators, and systematically summarizes the application of various nanoparticles in inducing cuproptosis, ranging from inorganic copper compounds to delivery systems. These nanoparticles offer significant advantages, such as improving copper absorption, extending the duration of effectiveness, enhancing the precision of copper release, increasing biocompatibility, and serving as enhancers in combination therapy. In conclusion, the authors present a detailed overview and insights into the current research directions of nanoplatforms that facilitate copper-induced cancer treatment, establishing a foundation for the future development of effective nanomedicines that induce cuproptosis and offering new possibilities and treatment strategies for tumor therapy.
Collapse
Affiliation(s)
- Yanqiang Yang
- Department of Respiratory Diseases, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
- Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, China
| | - Chen Dong
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| | - Xuehua Ma
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| | - Yanan Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| | - Zhouhua Li
- Department of Respiratory Diseases, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| | - Yuan Xu
- Department of Respiratory Diseases, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
- Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, China
| | - Tianxiang Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| | - Changyong Gao
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| | - Xiaoqun Ye
- Department of Respiratory Diseases, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
- Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, China
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| | - Xinyi Zhang
- Department of Respiratory Diseases, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
- Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, China
| |
Collapse
|
12
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2025; 480:759-784. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
13
|
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: 0] [Impact Index Per Article: 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
|
14
|
Yan X, Liu H, Guo L, Liu C, Zhang S, Wang X, Tang Y, Zhou R, Jiang X, Wang E, Gao S, Xu C. Multifunctional drug delivery nanoparticles for combined chemotherapy/chemodynamic/photothermal therapy against colorectal cancer through synergistic cuproptosis/ferroptosis/apoptosis. Mater Today Bio 2025; 30:101427. [PMID: 39850244 PMCID: PMC11754682 DOI: 10.1016/j.mtbio.2024.101427] [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: 10/30/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
The use of combination therapies that employ a variety of cell death mechanisms has emerged as a promising avenue of research in the treatment of cancer. However, the optimization of therapeutic synergies when integrating different modes remains a significant challenge. To this end, we developed a multifunctional intelligent drug-carrying nanoparticle (DFMTCH NPs) based on the metal-organic framework MIL-100, loaded with doxorubicin (DOX) and disulfiram (DSF), coated with a Cu-tannic acid (Cu-TA) network and hyaluronic acid (HA), for the purpose of combined chemotherapy/chemodynamic/photothermal anti-cancer therapy. On the one hand, the DFMTCH NPs exhibited a range of therapeutic capabilities, including chemotherapy, photothermal therapy (PTT), and chemodynamic therapy (CDT), which collectively enhanced the anti-tumor efficacy of chemotherapeutic agents. In addition, DFMTCH NPs proved sensitive photoacoustic imaging (PAI) in image-guided therapy. On the other hand, DFMTCH NPs could produce reactive oxygen species (ROS) and consume glutathione (GSH) by amplifying cellular oxidative stress, while causing intracellular mitochondrial dysfunction, inducing effective cuproptosis/ferroptosis/apoptosis to inhibit tumor growth. Collectively, this work provided an innovative strategy for designing multifunctional nanoparticles for effective combination therapies to combat colorectal cancer (CRC).
Collapse
Affiliation(s)
- Xiuzhang Yan
- Department of Gastrointestinal Colorectal Surgery, Jilin University China-Japan Union Hospital, Changchun, 130000, China
| | - Heshi Liu
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Lei Guo
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Chang Liu
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Shichen Zhang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xue Wang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yixin Tang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Rui Zhou
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xin Jiang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Erlei Wang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Shuohui Gao
- Department of Gastrointestinal Colorectal Surgery, Jilin University China-Japan Union Hospital, Changchun, 130000, China
| | - Caina Xu
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| |
Collapse
|
15
|
Imam M, Ji J, Zhang Z, Yan S. Targeting the initiator to activate both ferroptosis and cuproptosis for breast cancer treatment: progress and possibility for clinical application. Front Pharmacol 2025; 15:1493188. [PMID: 39867656 PMCID: PMC11757020 DOI: 10.3389/fphar.2024.1493188] [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: 09/08/2024] [Accepted: 11/12/2024] [Indexed: 01/28/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide. Metal metabolism is pivotal for regulating cell fate and drug sensitivity in breast cancer. Iron and copper are essential metal ions critical for maintaining cellular function. The accumulation of iron and copper ions triggers distinct cell death pathways, known as ferroptosis and cuproptosis, respectively. Ferroptosis is characterized by iron-dependent lipid peroxidation, while cuproptosis involves copper-induced oxidative stress. They are increasingly recognized as promising targets for the development of anticancer drugs. Recently, compelling evidence demonstrated that the interplay between ferroptosis and cuproptosis plays a crucial role in regulating breast cancer progression. This review elucidates the converging pathways of ferroptosis and cuproptosis in breast cancer. Moreover, we examined the value of genes associated with ferroptosis and cuproptosis in the clinical diagnosis and treatment of breast cancer, mainly outlining the potential for a co-targeting approach. Lastly, we delve into the current challenges and limitations of this strategy. In general, this review offers an overview of the interaction between ferroptosis and cuproptosis in breast cancer, offering valuable perspectives for further research and clinical treatment.
Collapse
Affiliation(s)
| | | | | | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Li L, Zhao J, Wang Y, Zhang Z, Chen W, Wang J, Cai Y. Integration of machine learning and experimental validation to identify the prognostic signature related to diverse programmed cell deaths in breast cancer. Front Oncol 2025; 14:1505934. [PMID: 39834939 PMCID: PMC11744720 DOI: 10.3389/fonc.2024.1505934] [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: 10/04/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Background Programmed cell death (PCD) is closely related to the occurrence, development, and treatment of breast cancer. The aim of this study was to investigate the association between various programmed cell death patterns and the prognosis of breast cancer (BRCA) patients. Methods The levels of 19 different programmed cell deaths in breast cancer were assessed by ssGSEA analysis, and these PCD scores were summed to obtain the PCDS for each sample. The relationship of PCDS with immune as well as metabolism-related pathways was explored. PCD-associated subtypes were obtained by unsupervised consensus clustering analysis, and differentially expressed genes between subtypes were analyzed. The prognostic signature (PCDRS) were constructed by the best combination of 101 machine learning algorithm combinations, and the C-index of PCDRS was compared with 30 published signatures. In addition, we analyzed PCDRS in relation to immune as well as therapeutic responses. The distribution of genes in different cells was explored by single-cell analysis and spatial transcriptome analysis. Potential drugs targeting key genes were analyzed by Cmap. Finally, the expression levels of key genes in clinical tissues were verified by RT-PCR. Results PCDS showed higher levels in cancer compared to normal. Different PCDS groups showed significant differences in immune and metabolism-related pathways. PCDRS, consisting of seven key genes, showed robust predictive ability over other signatures in different datasets. The high PCDRS group had a poorer prognosis and was strongly associated with a cancer-promoting tumor microenvironment. The low PCDRS group exhibited higher levels of anti-cancer immunity and responded better to immune checkpoint inhibitors as well as chemotherapy-related drugs. Clofibrate and imatinib could serve as potential small-molecule complexes targeting SLC7A5 and BCL2A1, respectively. The mRNA expression levels of seven genes were upregulated in clinical cancer tissues. Conclusion PCDRS can be used as a biomarker to assess the prognosis and treatment response of BRCA patients, which offers novel insights for prognostic monitoring and treatment personalization of BRCA patients.
Collapse
Affiliation(s)
- Longpeng Li
- Department of Anesthesiology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Jinfeng Zhao
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Yaxin Wang
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Zhibin Zhang
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Wanquan Chen
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Jirui Wang
- Institute of Physical Education and Sport, Shanxi University, Taiyuan, China
| | - Yue Cai
- Department of Anesthesiology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
17
|
Li T, Yu C. Metal-Dependent Cell Death in Renal Fibrosis: Now and in the Future. Int J Mol Sci 2024; 25:13279. [PMID: 39769044 PMCID: PMC11678559 DOI: 10.3390/ijms252413279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Renal fibrosis is a common final pathway underlying nearly almost all progressive kidney diseases. Metal ions are essential trace elements in organisms and are involved in important physiological activities. However, aberrations in intracellular metal ion metabolism may disrupt homeostasis, causing cell death and increasing susceptibility to various diseases. Accumulating evidence suggests a complex association between metal-dependent cell death and renal fibrosis. In this article, we provide a comprehensive overview of the specific molecular mechanisms of metal-dependent cell death and their crosstalk, up-to-date evidence supporting their role in renal fibrosis, therapeutic targeting strategies, and research needs, aiming to offer a rationale for future clinical treatment of renal fibrosis.
Collapse
Affiliation(s)
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
18
|
Huo G, Lin Y, Liu L, He Y, Qu Y, Liu Y, Zhu R, Wang B, Gong Q, Han Z, Yin H. Decoding ferroptosis: transforming orthopedic disease management. Front Pharmacol 2024; 15:1509172. [PMID: 39712490 PMCID: PMC11659002 DOI: 10.3389/fphar.2024.1509172] [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: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
As a mechanism of cell death, ferroptosis has gained popularity since 2012. The process is distinguished by iron toxicity and phospholipid accumulation, in contrast to autophagy, apoptosis, and other cell death mechanisms. It is implicated in the advancement of multiple diseases across the body. Researchers currently know that osteosarcoma, osteoporosis, and other orthopedic disorders are caused by NRF2, GPX4, and other ferroptosis star proteins. The effective relief of osteoarthritis symptoms from deterioration has been confirmed by clinical treatment with multiple ferroptosis inhibitors. At the same time, it should be reminded that the mechanisms involved in ferroptosis that regulate orthopedic diseases are not currently understood. In this manuscript, we present the discovery process of ferroptosis, the mechanisms involved in ferroptosis, and the role of ferroptosis in a variety of orthopedic diseases. We expect that this manuscript can provide a new perspective on clinical diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Guanlin Huo
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lusheng Liu
- Department of Acupuncture and Moxibustion, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqi He
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Yi Qu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yang Liu
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Renhe Zhu
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Bo Wang
- Department of Orthopaedics, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Qing Gong
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Zhongyu Han
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongbing Yin
- Orthopedic Center, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
19
|
Chen H, Tang T, Xue C, Liu X, Xi Z, Xie L, Kang R. Exploration and breakthrough in the mode of intervertebral disc cell death may lead to significant advances in treatments for intervertebral disc degeneration. J Orthop Surg Res 2024; 19:825. [PMID: 39639370 PMCID: PMC11619685 DOI: 10.1186/s13018-024-05280-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Low back pain caused by intervertebral disc degeneration (IDD) has emerged as a significant global public health concern, with far-reaching consequences for patients' quality of life and healthcare systems. Although previous research have revealed that the mechanisms of intervertebral disc cell apoptosis, pyroptosis and necroptosis can aggravate IDD damage by mediating inflammation and promoting extracellular matrix degradation, but they cannot explain the connection between different cell death mechanisms and ion metabolism disorders. The latest study shows that cell death mechanisms such as cellular senescence, ferroptosis, and cuproptosis, and PANopotosis have similar roles in the progression of intervertebral disc degeneration, but not exactly the same damage mechanism. This paper summarizes the effects of various cell death patterns on the disease progression of IDD, related molecular mechanisms and signaling pathways, providing new perspectives and potential clinical intervention strategies for the prevention and treatment of IDD.
Collapse
Affiliation(s)
- Heng Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Tian Tang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Congyang Xue
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Xin Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Zhipeng Xi
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Lin Xie
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
- Jiangsu Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| | - Ran Kang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
- Jiangsu Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
20
|
Hu J, Zhu J, Chen T, Zhao Y, Xu Q, Wang Y. Cuproptosis in cancer therapy: mechanisms, therapeutic application and future prospects. J Mater Chem B 2024; 12:12191-12206. [PMID: 39526989 DOI: 10.1039/d4tb01877j] [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: 11/16/2024]
Abstract
Cuproptosis is a regulated form of cell death induced by the accumulation of metal ions and is closely linked to aspects of cellular drug resistance, cellular metabolism, and signalling pathways. Due to its crucial role in regulating physiological and pathological processes, cuproptosis has gained increasing significance as a potential target for anticancer drug development. In this review, we introduce the definition of cuproptosis and provide a comprehensive discussion of the mechanisms of cuproptosis. In addition, the methods for the detection of cuproptosis are summarized, and recent advances in cuproptosis in cancer therapy are reviewed, mainly in terms of elesclomol (ES)-mediated cuproptosis and disulfiram (DSF)-mediated cuproptosis, which provided practical value for applications. Finally, the current challenges and future development of cuproptosis-mediated cancer therapy are discussed. In summary, this review highlights recent progress on cuproptosis in cancer therapy, offering novel ideas and strategies for future research and applications.
Collapse
Affiliation(s)
- Jiawei Hu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Junfei Zhu
- China-Japan Friendship Hospital, No. 2 Sakura East Street, Chaoyang District, Beijing, China
| | - Tao Chen
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yudie Zhao
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Qingwen Xu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| |
Collapse
|
21
|
Lu X, Deng W, Wang S, Zhao S, Zhu B, Bai B, Mao Y, Lin J, Yi Y, Xie Z, Wang X, Lu Y, Huang X, You T, Chen X, Sun W, Shen X. PEGylated Elesclomol@Cu(Ⅱ)-based Metal‒organic framework with effective nanozyme performance and cuproptosis induction efficacy for enhanced PD-L1-based immunotherapy. Mater Today Bio 2024; 29:101317. [PMID: 39554839 PMCID: PMC11565527 DOI: 10.1016/j.mtbio.2024.101317] [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: 08/15/2024] [Revised: 10/13/2024] [Accepted: 10/27/2024] [Indexed: 11/19/2024] Open
Abstract
Nanozymes constitute a promising treatment strategy for antitumor therapy. However, the catalytic function of metal‒organic framework (MOF)-based nanozymes during cuproptosis remains unclear. In this study, a Cu(Ⅱ)-based MOF nanocomposite loaded with the copper ionophore elesclomol and surface modified with polyethylene glycol polymer (PEG) was developed (ES@Cu(Ⅱ)-MOF) for effective cuproptosis induction. The peroxidase (POD)-like activity of ES@Cu(Ⅱ)-MOF generated an abundance of hydroxyl radicals (•OH) via a Fenton-like reaction, and its glutathione peroxidase (GSH-Px)-like activity converted Cu2+ into more toxic Cu+ ions to efficiently consume endogenous GSH. Notably, the rapid accumulation of Cu+ and ES in tumor cells induced the aggregation of lipoylated dihydrolipoamide S-acetyltransferase (DLAT) and the downregulation of Fe‒S cluster proteins, ultimately leading to cuproptosis. ES@Cu(Ⅱ)-MOF exhibited extraordinary cytotoxicity against breast cancer cells in vitro and significantly suppressed 4T1 breast tumor growth in vivo. Moreover, ES@Cu(Ⅱ)-MOF induced immunogenic cell death (ICD) to increase the antitumor immune response. Furthermore, combining ES@Cu(Ⅱ)-MOF with an anti-programmed cell death-ligand 1 (PD-L1) antibody converted the immunosuppressive tumor microenvironment to an immunogenic microenvironment, thus effectively inhibiting breast tumor growth. Overall, this work provides an innovative approach utilizing nanozymes to facilitate cuproptosis for cancer treatment, which potentially enhances the effectiveness of immune checkpoint inhibitor-based immunotherapy.
Collapse
Affiliation(s)
- Xufeng Lu
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wenhai Deng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Shuaibin Wang
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Shengsheng Zhao
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Bingzi Zhu
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Binglong Bai
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yiwen Mao
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ji Lin
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yongdong Yi
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Zuoliang Xie
- Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiang Wang
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yongyong Lu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiufeng Huang
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Tao You
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiaolei Chen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Weijian Sun
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xian Shen
- Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
22
|
Gromadzka G, Antos A, Sorysz Z, Litwin T. Psychiatric Symptoms in Wilson's Disease-Consequence of ATP7B Gene Mutations or Just Coincidence?-Possible Causal Cascades and Molecular Pathways. Int J Mol Sci 2024; 25:12354. [PMID: 39596417 PMCID: PMC11595239 DOI: 10.3390/ijms252212354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Wilson's disease (WD) is an autosomal recessive disorder of copper metabolism. The genetic defect in WD affects the ATP7B gene, which encodes the ATP7B transmembrane protein, which is essential for maintaining normal copper homeostasis in the body. It is primarily expressed in the liver and acts by incorporating copper into ceruloplasmin (Cp), the major copper transport protein in the blood. In conditions of excess copper, ATP7B transports it to bile for excretion. Mutations in ATP7B lead to impaired ATP7B function, resulting in copper accumulation in hepatocytes leading to their damage. The toxic "free"-unbound to Cp-copper released from hepatocytes then accumulates in various organs, contributing to their damage and clinical manifestations of WD, including hepatic, neurological, hematological, renal, musculoskeletal, ophthalmological, psychiatric, and other effects. While most clinical manifestations of WD correspond to identifiable organic or cellular damage, the pathophysiology underlying its psychiatric manifestations remains less clearly understood. A search for relevant articles was conducted in PubMed/Medline, Science Direct, Scopus, Willy Online Library, and Google Scholar, combining free text and MeSH terms using a wide range of synonyms and related terms, including "Wilson's disease", "hepatolenticular degeneration", "psychiatric manifestations", "molecular mechanisms", "pathomechanism", and others, as well as their combinations. Psychiatric symptoms of WD include cognitive disorders, personality and behavioral disorders, mood disorders, psychosis, and other mental disorders. They are not strictly related to the location of brain damage, therefore, the question arises whether these symptoms are caused by WD or are simply a coincidence or a reaction to the diagnosis of a genetic disease. Hypotheses regarding the etiology of psychiatric symptoms of WD suggest a variety of molecular mechanisms, including copper-induced CNS toxicity, oxidative stress, mitochondrial dysfunction, mitophagy, cuproptosis, ferroptosis, dysregulation of neurotransmission, deficiencies of neurotrophic factors, or immune dysregulation. New studies on the expression of noncoding RNA in WD are beginning to shed light on potential molecular pathways involved in psychiatric symptomatology. However, current evidence is still insufficient to definitively establish the cause of psychiatric symptoms in WD. It is possible that the etiology of psychiatric symptoms varies among individuals, with multiple biological and psychological mechanisms contributing to them simultaneously. Future studies with larger samples and comprehensive analyses are necessary to elucidate the mechanisms underlying the psychiatric manifestations of WD and to optimize diagnostics and therapeutic approaches.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Department of Biomedical Sciences, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University, Wóycickiego Street 1/3, 01-938 Warsaw, Poland
| | - Agnieszka Antos
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego Street 9, 02-957 Warsaw, Poland;
| | - Zofia Sorysz
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis Street 5, 01-815 Warsaw, Poland
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego Street 9, 02-957 Warsaw, Poland;
| |
Collapse
|
23
|
Shen Z, Qiu Y, Ding H, Ren F, Chen H. Cuproptosis and Cuproptosis-Based Synergistic Therapy for Cancer Treatment. ChemMedChem 2024; 19:e202400216. [PMID: 38943463 DOI: 10.1002/cmdc.202400216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/01/2024]
Abstract
Copper, as an essential trace nutrient for human, plays a crucial role in numerous cellular activities, and is vital for maintaining homeostasis in organisms. Deviations from normal intracellular copper concentration range can disrupt the cellular homeostasis and lead to cell death. Cell death is the process in which cells lose their vitality and cannot sustain normal metabolism, which has various forms. The recently discovered cuproptosis mechanism differs from the previously recognized forms, which is triggered by intracellular copper accumulation. The discovery of cuproptosis has sparked interest among researchers, and this mechanism has been applied in the treatment of various intractable diseases, including different types of cancer. However, the developed cuproptosis-based therapies have revealed certain limitations, such as low immunostimulatory efficiency, poor tumor targeting, and inhibition by the tumor microenvironment. Therefore, researchers are devoted to combining cuproptosis with existing cancer therapies to develop more effective synergistic cancer therapies. This review summarizes the latest research advancements in the cuproptosis-based therapies for various types of cancer, with a focus on the synergistic cancer therapies. Finally, it provides an outlook on the future development of cuproptosis in anti-tumor therapy.
Collapse
Affiliation(s)
- Zhiyang Shen
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, China
| | - Yu Qiu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, China
| | - Haizhen Ding
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, China
| | - Fangfang Ren
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, China
| | - Hongmin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
24
|
Zhang L, Xie A, Ma J, Liu H, Zeng C. Unveiling Cuproptosis: Mechanistic insights, roles, and leading advances in oncology. Biochim Biophys Acta Rev Cancer 2024; 1879:189180. [PMID: 39276875 DOI: 10.1016/j.bbcan.2024.189180] [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: 05/17/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
Copper, a vital micronutrient, performs essential functions in numerous biological settings. Its disrupted metabolism is implicated in both the initiation of tumors and therapeutic interventions for cancer, underscoring the critical necessity of preserving copper homeostasis. Cuproptosis, a regulated cell death (RCD) modulated by copper, is activated in response to elevated copper concentrations, prompting an investigation into its implication in oncogenesis. Within this review, an exploration is conducted into copper dynamics and homeostasis maintenance within cells. Furthermore, it delves into the mechanisms underlying cuproptosis and its interplay with signaling pathways implicated in cancer. The potential synergy between cuproptosis and ferroptosis and its impact on tumor immunomodulation is discussed. Additionally, promising avenues for addressing cuproptosis in cancer involve assessing the utility of copper chelators and ionophores. By addressing pressing questions surrounding cuproptosis and outlining its pivotal role in cancer pathogenesis and treatment, this review propounds targeting cuproptosis as a promising frontier in antitumor therapy, potentially revolutionizing cancer treatment strategies.
Collapse
Affiliation(s)
- Limei Zhang
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Aihui Xie
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Jingxian Ma
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Huilin Liu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen 518110, China.
| |
Collapse
|
25
|
Aquilano K, Filomeni G, Faraonio R, De Luca A. Editorial: Metals in cancer: from intracellular signaling to therapy. Front Oncol 2024; 14:1495825. [PMID: 39450254 PMCID: PMC11499201 DOI: 10.3389/fonc.2024.1495825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Affiliation(s)
- Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Redox Biology Research Group, Danish Cancer Institute, Copenhagen, Denmark
| | - Raffaella Faraonio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
26
|
Bhat AA, Afzal M, Moglad E, Thapa R, Ali H, Almalki WH, Kazmi I, Alzarea SI, Gupta G, Subramaniyan V. lncRNAs as prognostic markers and therapeutic targets in cuproptosis-mediated cancer. Clin Exp Med 2024; 24:226. [PMID: 39325172 PMCID: PMC11427524 DOI: 10.1007/s10238-024-01491-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as crucial regulators in various cellular processes, including cancer progression and stress response. Recent studies have demonstrated that copper accumulation induces a unique form of cell death known as cuproptosis, with lncRNAs playing a key role in regulating cuproptosis-associated pathways. These lncRNAs may trigger cell-specific responses to copper stress, presenting new opportunities as prognostic markers and therapeutic targets. This paper delves into the role of lncRNAs in cuproptosis-mediated cancer, underscoring their potential as biomarkers and targets for innovative therapeutic strategies. A thorough review of scientific literature was conducted, utilizing databases such as PubMed, Google Scholar, and ScienceDirect, with search terms like 'lncRNAs,' 'cuproptosis,' and 'cancer.' Studies were selected based on their relevance to lncRNA regulation of cuproptosis pathways and their implications for cancer prognosis and treatment. The review highlights the significant contribution of lncRNAs in regulating cuproptosis-related genes and pathways, impacting copper metabolism, mitochondrial stress responses, and apoptotic signaling. Specific lncRNAs are potential prognostic markers in breast, lung, liver, ovarian, pancreatic, and gastric cancers. The objective of this article is to explore the role of lncRNAs as potential prognostic markers and therapeutic targets in cancers mediated by cuproptosis.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
27
|
Lonardo A, Weiskirchen R. Copper and liver fibrosis in MASLD: the two-edged sword of copper deficiency and toxicity. METABOLISM AND TARGET ORGAN DAMAGE 2024. [DOI: 10.20517/mtod.2024.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Copper is a trace metal whose absence or deficiency can cause structural and functional alterations that can be corrected by copper administration. Copper excess is associated with significant liver toxicity, such as that seen in Wilson’s disease, which often exhibits liver steatosis and can be managed by copper sequestrants. Copper, due to its ability to either accept or donate electrons, is a cofactor in many physiological redox reactions, playing an essential role in cell energy homeostasis, detoxification of reactive oxygen species, and hepatic immunometabolism. Given these facts, it is reasonable to speculate that copper might be involved in the pathogenesis of liver fibrosis in the setting of metabolic dysfunction-associated fatty liver disease (MASLD). To address this research question, a narrative review of published studies was conducted, spanning from the needs, sources, and toxicity of copper to Menkes and Wilson’s disease. Most epidemiological studies have demonstrated that MASLD is associated with copper deficiency. However, several studies show that MASLD is associated with copper excess and very few conclude that copper is not associated with MASLD. Therefore, the putative pathomechanisms associating both copper excess and deficiency with MASLD development and progression are reviewed. In conclusion, epidemiological and pathogenic data support the notion that well-balanced copper homeostasis is a prerequisite for liver health. Accordingly, both copper excess and deficiency may potentially predispose to liver fibrosis via the development of MASLD. Therefore, studies aimed at restoring normal bodily stores of copper should be tailored according to precision medicine approaches based on the specific features of copper metabolism in individual MASLD patients.
Collapse
|
28
|
Kurmangaliyeva S, Baktikulova K, Tkachenko V, Seitkhanova B, Shapambayev N, Rakhimzhanova F, Almagambetova A, Kurmangaliyev K. An Overview of Hexavalent Chromium-Induced Necroptosis, Pyroptosis, and Ferroptosis. Biol Trace Elem Res 2024:10.1007/s12011-024-04376-1. [PMID: 39287767 DOI: 10.1007/s12011-024-04376-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Heavy metals are common environmental industrial pollutants. Due to anthropogenic activity, chromium, especially its hexavalent form [Cr(VI)], is a widespread environmental contaminant that poses a threat to human health. In this review paper, we summarize the currently reported molecular mechanisms involved in chromium toxicity with a focus on the induction of pro-inflammatory non-apoptotic cell death pathways such as necroptosis, pyroptosis, and ferroptosis. The review highlights the ability of chromium to induce necroptosis, pyroptosis, and ferroptosis revealing the signaling pathways involved. Cr(VI) can induce RIPK1/RIPK3-dependent necroptosis both in vitro and in vivo. Chromium toxicity is associated with pyroptotic NLRP3 inflammasome/caspase-1/gasdermin D-dependent secretion of IL-1β and IL-18. Furthermore, this review emphasizes the role of redox imbalance and intracellular iron accumulation in Cr(VI)-induced ferroptosis. Of note, the crosstalk between the investigated lethal subroutines in chromium-induced toxicity is primarily mediated by reactive oxygen species (ROS), which are suggested to act as a rheostat determining the cell death pathway in cells exposed to chromium. The current study provides novel insights into the pro-inflammatory effects of chromium, since necroptosis, pyroptosis, and ferroptosis affect inflammation owing to their immunogenic properties linked primarily with damage-associated molecular patterns. Inhibition of these non-apoptotic lethal subroutines can be considered a therapeutic strategy to reduce the toxicity of heavy metals, including chromium.
Collapse
Affiliation(s)
- Saulesh Kurmangaliyeva
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kristina Baktikulova
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan.
| | - Viktoriya Tkachenko
- State Institution "Republican Scientific and Practical Centre of Sports, " 8 Narochanskaya St, Minsk, Republic of Belarus
| | - Bibigul Seitkhanova
- Department of Microbiology, Virology and Immunology, South Kazakhstan Medical Academy, Al-Farabi Sq, Shymkent, Republic of Kazakhstan
| | - Nasriddin Shapambayev
- Department of General Practitioner - 1, Khoja Akhmet Yasawi International Kazakh-Turkish University, 7/7 Baitursynov St, Shymkent, Republic of Kazakhstan
| | - Farida Rakhimzhanova
- Department of Microbiology, NCJSC "Semey Medical University, " 103 Abay St, Semey, Republic of Kazakhstan
| | - Altyn Almagambetova
- Department of Phthisiology and Dermatovenerology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kairat Kurmangaliyev
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| |
Collapse
|
29
|
Wang SH, Hsieh YY, Ong KH, Lai HY, Tsai HH, Sun DP, Huang SKH, Tian YF, Wu HC, Chan TC, Joseph K, Chang IW. The clinicopathological significance and prognostic impact of 14-3-3σ/stratifin expression on patients with surgically resectable intrahepatic cholangiocarcinoma. Asian J Surg 2024:S1015-9584(24)01873-6. [PMID: 39232956 DOI: 10.1016/j.asjsur.2024.08.133] [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: 05/01/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer after hepatocellular carcinoma. Through data mining of publicly available iCCA transcriptomic datasets from the Gene Expression Omnibus, we identified SFN as the most significantly up-regulated gene in iCCA compared to normal tissue, focusing on the Gene Ontology term "cell proliferation" (GO:0008283). SFN encodes the 14-3-3σ protein, also known as stratifin, which plays crucial roles in various cellular processes. MATERIALS AND METHODS Immunohistochemistry was used to assess stratifin expression in 182 patients with localized iCCAs undergoing surgical resection. Patients were divided into low and high expression groups, and the association between stratifin expression and clinicopathological features was analyzed. Univariate and multivariate survival analyses were performed to assess overall survival (OS), disease-specific survival (DSS), local recurrence-free survival (LRFS), and metastasis-free survival (MeFS). RESULTS Elevated stratifin expression in iCCAs was significantly associated with the absence of hepatitis, positive surgical margins, advanced primary tumor stages, and higher histological grades (all p ≤ 0.011). Survival analyses demonstrated a significant negative association between stratifin expression and all prognostic indicators, including OS, DSS, LRFS, and MeFS (all p ≤ 0.0004). Multivariate analysis revealed that stratifin overexpression was significantly correlated with poorer outcomes in terms of DSS, LRFS, and MeFS (all p < 0.001). CONCLUSIONS These findings suggest that stratifin may play a crucial role in iCCA oncogenesis and tumor progression, serving as a potential novel prognostic biomarker.
Collapse
Affiliation(s)
- Su-Hong Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Yao-Yu Hsieh
- Division of Hematology and Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan; Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Khaa Hoo Ong
- Department of Surgery, Division of Gastroenterology and General Surgery, Chi Mei Medical Center, Tainan, Taiwan; Department of Medical Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hong-Yue Lai
- Department of Pharmacology, School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hsin-Hwa Tsai
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ding-Ping Sun
- Department of Surgery, Division of Gastroenterology and General Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Steven Kuan-Hua Huang
- Department of Surgery, Division of Urology, Chi Mei Medical Center, Tainan, Taiwan; Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Yu-Feng Tian
- Department of Surgery, Division of Colon and Rectal Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Hung-Chang Wu
- Department of Internal Medicine, Division of Hematology and Oncology, Chi Mei Medical Center, Tainan, Taiwan; College of Pharmacy and Science, Chia Nan University, Tainan, Taiwan
| | - Ti-Chun Chan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | | | - I-Wei Chang
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Clinical Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Pathology, Taipei Medical University Hospital, Taipei, Taiwan; Department of Pathology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
30
|
Zhou Z, Mai Y, Zhang G, Wang Y, Sun P, Jing Z, Li Z, Xu Y, Han B, Liu J. Emerging role of immunogenic cell death in cancer immunotherapy: Advancing next-generation CAR-T cell immunotherapy by combination. Cancer Lett 2024; 598:217079. [PMID: 38936505 DOI: 10.1016/j.canlet.2024.217079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Immunogenic cell death (ICD) is a stress-driven form of regulated cell death (RCD) in which dying tumor cells' specific signaling pathways are activated to release damage-associated molecular patterns (DAMPs), leading to the robust anti-tumor immune response as well as a reversal of the tumor immune microenvironment from "cold" to "hot". Chimeric antigen receptor (CAR)-T cell therapy, as a landmark in anti-tumor immunotherapy, plays a formidable role in hematologic malignancies but falls short in solid tumors. The Gordian knot of CAR-T cells for solid tumors includes but is not limited to, tumor antigen heterogeneity or absence, physical and immune barriers of tumors. The combination of ICD induction therapy and CAR-T cell immunotherapy is expected to promote the intensive use of CAR-T cell in solid tumors. In this review, we summarize the characteristics of ICD, stress-responsive mechanism, and the synergistic effect of various ICD-based therapies with CAR-T cells to effectively improve anti-tumor capacity.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yumiao Mai
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Henan Province Key Laboratory of Cardiac Injury and Repair, Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, 450052, China
| | - Yingjie Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Pan Sun
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhaohe Jing
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jian Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
31
|
Gromadzka G, Czerwińska J, Krzemińska E, Przybyłkowski A, Litwin T. Wilson's Disease-Crossroads of Genetics, Inflammation and Immunity/Autoimmunity: Clinical and Molecular Issues. Int J Mol Sci 2024; 25:9034. [PMID: 39201720 PMCID: PMC11354778 DOI: 10.3390/ijms25169034] [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: 07/19/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Wilson's disease (WD) is a rare, autosomal recessive disorder of copper metabolism caused by pathogenic mutations in the ATP7B gene. Cellular copper overload is associated with impaired iron metabolism. Oxidative stress, cuproptosis, and ferroptosis are involved in cell death in WD. The clinical picture of WD is variable. Hepatic/neuropsychiatric/other symptoms may manifest in childhood/adulthood and even old age. It has been shown that phenotypic variability may be determined by the type of ATP7B genetic variants as well as the influence of various genetic/epigenetic, environmental, and lifestyle modifiers. In 1976, immunological abnormalities were first described in patients with WD. These included an increase in IgG and IgM levels and a decrease in the percentage of T lymphocytes, as well as a weakening of their bactericidal effect. Over the following years, it was shown that there is a bidirectional relationship between copper and inflammation. Changes in serum cytokine concentrations and the relationship between cytokine gene variants and the clinical course of the disease have been described in WD patients, as well as in animal models of this disease. Data have also been published on the occurrence of antinuclear antibodies (ANAs), antineutrophil cytoplasmic antibodies (ANCAs), anti-muscle-specific tyrosine kinase antibodies, and anti-acetylcholine receptor antibodies, as well as various autoimmune diseases, including systemic lupus erythematosus (SLE), myasthenic syndrome, ulcerative colitis, multiple sclerosis (MS), polyarthritis, and psoriasis after treatment with d-penicillamine (DPA). The occurrence of autoantibodies was also described, the presence of which was not related to the type of treatment or the form of the disease (hepatic vs. neuropsychiatric). The mechanisms responsible for the occurrence of autoantibodies in patients with WD are not known. It has also not been clarified whether they have clinical significance. In some patients, WD was differentiated or coexisted with an autoimmune disease, including autoimmune hepatitis or multiple sclerosis. Various molecular mechanisms may be responsible for immunological abnormalities and/or the inflammatory processes in WD. Their better understanding may be important for explaining the reasons for the diversity of symptoms and the varied course and response to therapy, as well as for the development of new treatment regimens for WD.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Department of Biomedical Sciences, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University, Wóycickiego Street 1/3, 01-938 Warsaw, Poland
| | - Julia Czerwińska
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis Street 5, 01-815 Warsaw, Poland
| | - Elżbieta Krzemińska
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis Street 5, 01-815 Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland;
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego Street 9, 02-957 Warsaw, Poland;
| |
Collapse
|
32
|
Hao S, Yang Z, Wang G, Cai G, Qin Y. Development of prognostic model incorporating a ferroptosis/cuproptosis-related signature and mutational landscape analysis in muscle-invasive bladder cancer. BMC Cancer 2024; 24:958. [PMID: 39107713 PMCID: PMC11302292 DOI: 10.1186/s12885-024-12741-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Muscle-invasive bladder cancer (MIBC) is a prevalent and aggressive malignancy. Ferroptosis and cuproptosis are recently discovered forms of programmed cell death (PCD) that have attracted much attention. However, their interactions and impacts on MIBC overall survival (OS) and treatment outcomes remain unclear. METHODS Data from the TCGA-BLCA project (as the training set), cBioPortal database, and GEO datasets (GSE13507 and GSE32894, as the test sets) were utilized to identify hub ferroptosis/cuproptosis-related genes (FRGs and CRGs) and develop a prognostic signature. Differential expression analysis (DEA) was conducted, followed by univariate and multivariate Cox's regression analyses and multiple machine learning (ML) techniques to select genetic features. The performance of the ferroptosis/cuproptosis-related signature was evaluated using Kaplan-Meier (K-M) survival analysis and receiver-operating characteristics (ROC) curves. Mutational and tumour immune microenvironment landscapes were also explored. Real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) experiments confirmed the expression patterns of the hub genes, and functional assays assessed the effects of SCD knockdown on cell viability, proliferation, and migration. RESULTS DEA revealed dysregulated FRGs and CRGs in the TCGA MIBC cohort. SCD, DDR2, and MT1A were identified as hub genes. A prognostic signature based on the sum of the weighted expression of these genes demonstrated strong predictive efficacy in the training and test sets. Nomogram incorporating this signature accurately predicted 1-, 3-, and 5-year survival probabilities in the TCGA cohort and GSE13507 dataset. Copy number variation (CNV) and tumour immune microenvironment analysis revealed that high risk score level groups were associated with immunosuppression and lower tumour purity. The associations of risk scores with immunotherapy and chemical drugs were also explored, indicating their potential for guiding treatment for MIBC patients. The dysregulated expression patterns of three hub genes were validated by RT-qPCR experiments. CONCLUSIONS Targeting hub FRGs and CRGs could be a promising therapeutic approach for MIBC. Our prognostic model offers a new framework for MIBC subtyping and can inform personalized therapeutic strategies.
Collapse
Affiliation(s)
- Sida Hao
- Department of Urology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310003Zhejiang , China
| | - Zitong Yang
- Department of Urology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Gang Wang
- Department of Urology, Affiliated Hangzhou First People's Hospital, Xihu University School of Medicine, Hangzhou, Zhejiang, China
| | - Guofeng Cai
- Department of Urology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310003Zhejiang , China
| | - Yong Qin
- Department of Urology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310003Zhejiang , China.
| |
Collapse
|
33
|
Yang Z, Feng R, Zhao H. Cuproptosis and Cu: a new paradigm in cellular death and their role in non-cancerous diseases. Apoptosis 2024:10.1007/s10495-024-01993-y. [PMID: 39014119 DOI: 10.1007/s10495-024-01993-y] [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] [Accepted: 06/24/2024] [Indexed: 07/18/2024]
Abstract
Cuproptosis, a newly characterized form of regulated cell death driven by copper accumulation, has emerged as a significant mechanism underlying various non-cancerous diseases. This review delves into the complex interplay between copper metabolism and the pathogenesis of conditions such as Wilson's disease (WD), neurodegenerative disorders, and cardiovascular pathologies. We examine the molecular mechanisms by which copper dysregulation induces cuproptosis, highlighting the pivotal roles of key copper transporters and enzymes. Additionally, we evaluate the therapeutic potential of copper chelation strategies, which have shown promise in experimental models by mitigating copper-induced cellular damage and restoring physiological homeostasis. Through a comprehensive synthesis of recent advancements and current knowledge, this review underscores the necessity of further research to translate these findings into clinical applications. The ultimate goal is to harness the therapeutic potential of targeting cuproptosis, thereby improving disease management and patient outcomes in non-cancerous conditions associated with copper dysregulation.
Collapse
Affiliation(s)
- Zhibo Yang
- Department of Neurosurgery, 3201 Hospital of Xi'an Jiaotong University Health Science Center, Hanzhong, 723000, Shaanxi, China
| | - Ridong Feng
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine (FAHZU), 79 Qingchun Rd., Shangcheng District, Hangzhou, 330100, Zhejiang, China
| | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China.
| |
Collapse
|
34
|
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
|
35
|
Zhang X, Han X. Targeting cuproptosis for cancer therapy: Focus on the anti-tumor immune system. CANCER PATHOGENESIS AND THERAPY 2024. [DOI: 10.1016/j.cpt.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
36
|
Kciuk M, Gielecińska A, Kałuzińska-Kołat Ż, Yahya EB, Kontek R. Ferroptosis and cuproptosis: Metal-dependent cell death pathways activated in response to classical chemotherapy - Significance for cancer treatment? Biochim Biophys Acta Rev Cancer 2024; 1879:189124. [PMID: 38801962 DOI: 10.1016/j.bbcan.2024.189124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Apoptosis has traditionally been regarded as the desired cell death pathway activated by chemotherapeutic drugs due to its controlled and non-inflammatory nature. However, recent discoveries of alternative cell death pathways have paved the way for immune-stimulatory treatment approaches in cancer. Ferroptosis (dependent on iron) and cuproptosis (dependent on copper) hold promise for selective cancer cell targeting and overcoming drug resistance. Copper ionophores and iron-bearing nano-drugs show potential for clinical therapy as single agents and as adjuvant treatments. Here we review up-to-date evidence for the involvement of metal ion-dependent cell death pathways in the cytotoxicity of classical chemotherapeutic agents (alkylating agents, topoisomerase inhibitors, antimetabolites, and mitotic spindle inhibitors) and their combinations with cuproptosis and ferroptosis inducers, indicating the prospects, advantages, and obstacles of their use.
Collapse
Affiliation(s)
- M Kciuk
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Biotechnology and Genetics, Banacha St. 12/16, 90-237 Lodz, Poland.
| | - A Gielecińska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Biotechnology and Genetics, Banacha St. 12/16, 90-237 Lodz, Poland; University of Lodz, Doctoral School of Exact and Natural Sciences, Banacha Street 12/16, 90-237 Lodz, Poland
| | - Ż Kałuzińska-Kołat
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - E B Yahya
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - R Kontek
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Biotechnology and Genetics, Banacha St. 12/16, 90-237 Lodz, Poland
| |
Collapse
|
37
|
Neagu AN, Jayaweera T, Weraduwage K, Darie CC. A Nanorobotics-Based Approach of Breast Cancer in the Nanotechnology Era. Int J Mol Sci 2024; 25:4981. [PMID: 38732200 PMCID: PMC11084175 DOI: 10.3390/ijms25094981] [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] [Revised: 04/28/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
We are living in an era of advanced nanoscience and nanotechnology. Numerous nanomaterials, culminating in nanorobots, have demonstrated ingenious applications in biomedicine, including breast cancer (BC) nano-theranostics. To solve the complicated problem of BC heterogeneity, non-targeted drug distribution, invasive diagnostics or surgery, resistance to classic onco-therapies and real-time monitoring of tumors, nanorobots are designed to perform multiple tasks at a small scale, even at the organelles or molecular level. Over the last few years, most nanorobots have been bioengineered as biomimetic and biocompatible nano(bio)structures, resembling different organisms and cells, such as urchin, spider, octopus, fish, spermatozoon, flagellar bacterium or helicoidal cyanobacterium. In this review, readers will be able to deepen their knowledge of the structure, behavior and role of several types of nanorobots, among other nanomaterials, in BC theranostics. We summarized here the characteristics of many functionalized nanodevices designed to counteract the main neoplastic hallmark features of BC, from sustaining proliferation and evading anti-growth signaling and resisting programmed cell death to inducing angiogenesis, activating invasion and metastasis, preventing genomic instability, avoiding immune destruction and deregulating autophagy. Most of these nanorobots function as targeted and self-propelled smart nano-carriers or nano-drug delivery systems (nano-DDSs), enhancing the efficiency and safety of chemo-, radio- or photodynamic therapy, or the current imagistic techniques used in BC diagnosis. Most of these nanorobots have been tested in vitro, using various BC cell lines, as well as in vivo, mainly based on mice models. We are still waiting for nanorobots that are low-cost, as well as for a wider transition of these favorable effects from laboratory to clinical practice.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania;
| | - Taniya Jayaweera
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (T.J.); (K.W.)
| | - Krishan Weraduwage
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (T.J.); (K.W.)
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (T.J.); (K.W.)
| |
Collapse
|
38
|
Feng Y, Yang Z, Wang J, Zhao H. Cuproptosis: unveiling a new frontier in cancer biology and therapeutics. Cell Commun Signal 2024; 22:249. [PMID: 38693584 PMCID: PMC11064406 DOI: 10.1186/s12964-024-01625-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024] Open
Abstract
Copper plays vital roles in numerous cellular processes and its imbalance can lead to oxidative stress and dysfunction. Recent research has unveiled a unique form of copper-induced cell death, termed cuproptosis, which differs from known cell death mechanisms. This process involves the interaction of copper with lipoylated tricarboxylic acid cycle enzymes, causing protein aggregation and cell death. Recently, a growing number of studies have explored the link between cuproptosis and cancer development. This review comprehensively examines the systemic and cellular metabolism of copper, including tumor-related signaling pathways influenced by copper. It delves into the discovery and mechanisms of cuproptosis and its connection to various cancers. Additionally, the review suggests potential cancer treatments using copper ionophores that induce cuproptosis, in combination with small molecule drugs, for precision therapy in specific cancer types.
Collapse
Affiliation(s)
- Ying Feng
- Department of Emergency, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China
| | - Zhibo Yang
- Department of Neurosurgery, 3201 Hospital of Xi'an Jiaotong University Health Science Center, Hanzhong, 723000, Shaanxi, China
| | - Jianpeng Wang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China.
| |
Collapse
|
39
|
Li YH, Wang K. Editorial Commentary: Copper Homeostasis in Neurodegenerative Diseases. Curr Med Sci 2024; 44:244-245. [PMID: 38393531 DOI: 10.1007/s11596-024-2841-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Affiliation(s)
- Ying-Hui Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Kun Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|