1
|
Wu P, Chen J, Li H, Lu H, Li Y, Zhang J. Interactions between ferroptosis and tumour development mechanisms: Implications for gynaecological cancer therapy (Review). Oncol Rep 2025; 53:18. [PMID: 39635847 PMCID: PMC11638741 DOI: 10.3892/or.2024.8851] [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: 09/19/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Ferroptosis is a form of programmed cell death that is distinct from apoptosis. The mechanism involves redox‑active metallic iron and is characterized by an abnormal increase in iron‑dependent lipid reactive oxygen species, which results in high levels of membrane lipid peroxides. The relationship between ferroptosis and gynaecological tumours is complex. Ferroptosis can regulate tumour proliferation, metastasis and chemotherapy resistance, and targeting ferroptosis is a promising antitumour approach. Ferroptosis interacts with mechanisms related to tumorigenesis and development, such as macrophage polarization, the neutrophil trap network, mitochondrial autophagy and cuproptosis. The present review examines recent information on the interaction between the molecular mechanism of ferroptosis and other tumour‑related mechanisms, as well as the involvement of ferroptosis in gynaecological tumours, to identify implications for gynaecological cancer therapy.
Collapse
Affiliation(s)
- Peiting Wu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Jianlin Chen
- Department of Assisted Reproductive Centre, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Haiyuan Lu
- Department of Clinical Laboratory Department, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Hunan Vigorzoe Biotechnology Co., Ltd., Hunan 417700, P.R. China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| |
Collapse
|
2
|
Zhang Q, Zhang Y, Guo S, Wang H. Emerging insights into the role of microRNAs regulation of ferroptosis in hepatocellular carcinoma. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167642. [PMID: 39734007 DOI: 10.1016/j.bbadis.2024.167642] [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: 09/08/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Hepatocellular carcinoma (HCC) is a major type of liver cancer and an important cause of cancer death. It has been reported that the hepatocyte death plays an important role in HCC. Ferroptosis is an iron-dependent programmed cell death characterized by the accumulation of free iron and lipid peroxidation. A series of studies have shown that ferroptosis contributes to the occurrence and development of HCC. MicroRNAs (miRNAs) are non-coding RNAs with a length of approximately 21-22 nt. In recent years, miRNAs have been shown to participate in regulating ferroptosis to play a vital role in HCC, but the related mechanisms are not fully understood. This review summarized the current understanding of ferroptosis, as well as the biogenesis and function of miRNAs, and focused on the role of miRNAs regulation of ferroptosis in HCC, with the hope of providing new targets and ideas for the treatment of HCC.
Collapse
Affiliation(s)
- Qi Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yingdan Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
3
|
Zhang X, Xu W, Wang Z, Liu J, Gong H, Zou W. Cross-talk between cuproptosis and ferroptosis to identify immune landscape in cervical cancer for mRNA vaccines development. Eur J Med Res 2024; 29:602. [PMID: 39696618 DOI: 10.1186/s40001-024-02191-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
Messenger RNA (mRNA)-based vaccines present a promising avenue for cancer immunotherapy; however, their application in cervical cancer remains unexplored. This study investigated the interplay between the regulated cell death pathways of cuproptosis and ferroptosis to advance the development of mRNA vaccines for cervical cancer. We identified key cuproptosis-related and ferroptosis-related genes (CFRGs) from public mRNA profiles and determined their prognostic significance, mutation frequencies, and effect on the immune landscape. Our analysis revealed two distinct subtypes of cervical cancer associated with CFRGs, with differences in prognosis and immune characteristics. Using LASSO, XGBoost, and SVM-RFE methods, we established a 4-gene prognostic signature (TSC22D3, SQLE, ZNF419, and TFRC) to stratify patients based on their risk and determine its correlation with immune microenvironment, mutation profiles, and treatment responses. RT-qPCR validation confirmed the differential expression of these genes in clinical samples. Our findings identify TSC22D3, SQLE, ZNF419, and TFRC as candidate targets for mRNA vaccine development and offer a potential prognostic tool for personalized cervical cancer treatment.
Collapse
Affiliation(s)
- Xuchao Zhang
- Department of Oncology, The Second Xiangya Hospital, Central South University, No. 72 Xiangya Road, Changsha, 410000, Hunan, China
- Department of HematologyMolecular Biology Research Center, Center for Medical Genetics, School of Life SciencesHunan Province Key Laboratory of Basic and Applied Hematology, The Second Xiangya Hospital of Central South University, Central South University, No. 72 Xiangya Road, Changsha, 410011, China
| | - Wenwen Xu
- Department of Oncology, The Second Xiangya Hospital, Central South University, No. 72 Xiangya Road, Changsha, 410000, Hunan, China
- Department of HematologyMolecular Biology Research Center, Center for Medical Genetics, School of Life SciencesHunan Province Key Laboratory of Basic and Applied Hematology, The Second Xiangya Hospital of Central South University, Central South University, No. 72 Xiangya Road, Changsha, 410011, China
| | - Zi Wang
- Department of HematologyMolecular Biology Research Center, Center for Medical Genetics, School of Life SciencesHunan Province Key Laboratory of Basic and Applied Hematology, The Second Xiangya Hospital of Central South University, Central South University, No. 72 Xiangya Road, Changsha, 410011, China
| | - Jing Liu
- Department of HematologyMolecular Biology Research Center, Center for Medical Genetics, School of Life SciencesHunan Province Key Laboratory of Basic and Applied Hematology, The Second Xiangya Hospital of Central South University, Central South University, No. 72 Xiangya Road, Changsha, 410011, China
| | - Han Gong
- Department of Oncology, The Second Xiangya Hospital, Central South University, No. 72 Xiangya Road, Changsha, 410000, Hunan, China.
- Department of HematologyMolecular Biology Research Center, Center for Medical Genetics, School of Life SciencesHunan Province Key Laboratory of Basic and Applied Hematology, The Second Xiangya Hospital of Central South University, Central South University, No. 72 Xiangya Road, Changsha, 410011, China.
| | - Wen Zou
- Department of Oncology, The Second Xiangya Hospital, Central South University, No. 72 Xiangya Road, Changsha, 410000, Hunan, China.
| |
Collapse
|
4
|
Fan Q, He Y, Liu J, Liu Q, Wu Y, Chen Y, Dou Q, Shi J, Kong Q, Ou Y, Guo J. Large Language Model-Assisted Genotoxic Metal-Phenolic Nanoplatform for Osteosarcoma Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2403044. [PMID: 39670697 DOI: 10.1002/smll.202403044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Osteosarcoma, a leading primary bone malignancy in children and adolescents, is associated with a poor prognosis and a low global fertility rate. A large language model-assisted phenolic network (LLMPN) platform is demonstrated that integrates the large language model (LLM) GPT-4 into the design of multifunctional metal-phenolic network materials. Fine-tuned GPT-4 identified gossypol as a phenolic compound with superior efficacy against osteosarcoma after evaluating across a library of 60 polyphenols based on the correlation between experimental anti-osteosarcoma activity and multiplexed chemical properties of polyphenols. Subsequently, gossypol is then self-assembled into Cu2+-gossypol nanocomplexes with a hyaluronic acid surface modification (CuGOS NPs). CuGOS NPs has demonstrated the ability to induce genetic alterations and cell death in osteosarcoma cells, offering significant therapeutic benefits for primary osteosarcoma tumors and reducing metastasis without adverse effects on major organs or the genital system. This work presents an LLM-driven approach for engineering metal-organic nanoplatform and broadening applications by harnessing the capabilities of LLMs, thereby improving the feasibility and efficiency of research activities.
Collapse
Affiliation(s)
- Qingxin Fan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jialing Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qinling Liu
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yue Wu
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuxing Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China
| | - Qingyu Dou
- National Clinical Research Center for Geriatrics, Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Shi
- Section of Science and Education, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region (Hospital.C.T.), Chengdu, Sichuan, 610041, China
| | - Qingquan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Section of Science and Education, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region (Hospital.C.T.), Chengdu, Sichuan, 610041, China
| | - Yunsheng Ou
- Department of Orthopedics Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Departments of Chemical, Biological Engineering, The University of British Columbia, Vancouver, BC, V6T1Z4, Canada
| |
Collapse
|
5
|
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
|
6
|
Cui Y, Du X, Li Y, Wang D, Lv Z, Yuan H, Chen Y, Liu J, Sun Y, Wang W. Imbalanced and Unchecked: The Role of Metal Dyshomeostasis in Driving COPD Progression. COPD 2024; 21:2322605. [PMID: 38591165 DOI: 10.1080/15412555.2024.2322605] [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: 10/25/2023] [Accepted: 02/19/2024] [Indexed: 04/10/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic respiratory condition characterized by persistent inflammation and oxidative stress, which ultimately leads to progressive restriction of airflow. Extensive research findings have cogently suggested that the dysregulation of essential transition metal ions, notably iron, copper, and zinc, stands as a critical nexus in the perpetuation of inflammatory processes and oxidative damage within the lungs of COPD patients. Unraveling the intricate interplay between metal homeostasis, oxidative stress, and inflammatory signaling is of paramount importance in unraveling the intricacies of COPD pathogenesis. This comprehensive review aims to examine the current literature on the sources, regulation, and mechanisms by which metal dyshomeostasis contributes to COPD progression. We specifically focus on iron, copper, and zinc, given their well-characterized roles in orchestrating cytokine production, immune cell function, antioxidant depletion, and matrix remodeling. Despite the limited number of clinical trials investigating metal modulation in COPD, the advent of emerging methodologies tailored to monitor metal fluxes and gauge responses to chelation and supplementation hold great promise in unlocking the potential of metal-based interventions. We conclude that targeted restoration of metal homeostasis represents a promising frontier for ameliorating pathological processes driving COPD progression.
Collapse
Affiliation(s)
- Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xinqian Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yunqi Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Dan Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Huihui Yuan
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Jie Liu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ying Sun
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
7
|
Zhang Y, Zhang N, Xing J, Sun Y, Jin X, Shen C, Cheng L, Wang Y, Wang X. In situ hydrogel based on Cu-Fe 3O 4 nanoclusters exploits oxidative stress and the ferroptosis/cuproptosis pathway for chemodynamic therapy. Biomaterials 2024; 311:122675. [PMID: 38943822 DOI: 10.1016/j.biomaterials.2024.122675] [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: 04/08/2024] [Revised: 06/07/2024] [Accepted: 06/15/2024] [Indexed: 07/01/2024]
Abstract
Chemodynamic therapy (CDT) involving the use of metal nanozymes presents new opportunities for the treatment of deep-seated tumors. However, the lower ROS catalytic rate and dependence on high H2O2 concentrations affect therapeutic efficacy. To address this issue, a hydrogel was constructed for the treatment of osteosarcoma by combining Cu-Fe3O4 nanozymes (NCs) and artemisinin (AS) coencapsulated in situ with sodium alginate (ALG) and calcium ions. This hydrogel can release nanoparticles and AS within tumor tissue for an extended period of time, utilizing the multienzyme activity of NCs to achieve ROS accumulation. The carbon radicals (•C) generated from the interaction of Fe2+/Cu2+ with AS amplify oxidative stress, leading to tumor cell damage. Simultaneously, the NCs activate ferroptosis via the GPX4 pathway by depleting GSH and activate cuproptosis via the DLAT pathway by causing intracellular copper overload, enhancing therapeutic efficacy. In vitro experiments confirmed that the NCs-AS-ALG hydrogel has an excellent tumor cell killing effect, while in vivo experimental results demonstrated that it can effectively eliminate tumors with excellent biocompatibility, providing a new approach for osteosarcoma treatment.
Collapse
Affiliation(s)
- Yiqun Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China; College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, PR China
| | - Ni Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Jianghao Xing
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Yiwei Sun
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Xu Jin
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China
| | - Cailiang Shen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, PR China
| | - Yuanyin Wang
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, PR China.
| | - Xianwen Wang
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, PR China; School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, PR China.
| |
Collapse
|
8
|
Wu M, Li A, Zhang T, Ding W, Wei Y, Wan C, Ke B, Cheng H, Jin C, Kong C. The novel prognostic analysis of AML based on ferroptosis and cuproptosis related genes. J Trace Elem Med Biol 2024; 86:127517. [PMID: 39270538 DOI: 10.1016/j.jtemb.2024.127517] [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: 05/31/2024] [Revised: 07/27/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological malignancy. The aim of this research was to develop a ferroptosis and cuproptosis related novel prognostic signature associated with AML. METHODS The ferroptosis and cuproptosis related genes correlated with the prognosis of AML were identified by univariate Cox analysis. The consistent cluster analysis was performed for 150 AML patients in TCGA dataset. The key module genes associated with GSVA score of ferroptosis and cuproptosis were identified by WGCNA. univariate Cox and LASSO regression analysis were adopted to build a ferroptosis and cuproptosis AML prognostic signature. Finally, the expression of five prognostic genes in clinical tissue samples were verified by RT-qPCR. RESULTS A grand total of 27 FCRGs associated with AML prognosis were identified.Then, two AML sub-types with significantly different survival were obtained. We found 3 significantly differential expressed immune cells (naive CD4 cells, regulatory T cells and resting mast cells) between two risk sub-groups. Meanwhile, 'IL6 JAK STAT3 signaling' and 'P53 pathway' were enriched in low-risk group. A ferroptosis and cuproptosis related prognostic signature was build based on 8 prognostic genes. RT-qPCR results indicated that there was no significant difference in the expression of OLFML2A and CD109 between AML and normal samples. However, compared to the control group, LGALS1, SOCS1, and RHOC showed significantly lower expression in the AML group. CONCLUSION The prognostic signature comprised of OLFML2A, LGALS1, ABCB11, SOCS1, RHOC, CD109, RD3L and PTPN13 based on ferroptosis and cuproptosis was established, which provided theoretical basis for the research of AML.
Collapse
Affiliation(s)
- Mei Wu
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Anan Li
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Tingting Zhang
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Weirong Ding
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Yujing Wei
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Caishui Wan
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Bo Ke
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Hongbo Cheng
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Chenghao Jin
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Chunfang Kong
- Department of Hematology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China; Jiangxi Province Key Laboratory of Hematologic Diseases, Nanchang 330006, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Soochow 215006, China.
| |
Collapse
|
9
|
Alrouji M, Anwar S, Venkatesan K, Shahwan M, Hassan MI, Islam A, Shamsi A. Iron homeostasis and neurodegeneration in the ageing brain: Insight into ferroptosis pathways. Ageing Res Rev 2024; 102:102575. [PMID: 39515619 DOI: 10.1016/j.arr.2024.102575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/25/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Ageing is a major risk factor for various chronic diseases and offers a potential target for developing novel and broadly effective preventatives or therapeutics for age-related conditions, including those affecting the brain. Mechanisms contributing to ageing have been summarized as the hallmarks of ageing, with iron imbalance being one of the major factors. Ferroptosis, an iron-mediated lipid peroxidation-induced programmed cell death, has recently been implicated in neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Addressing ferroptosis offers both opportunities and challenges for treating neurodegenerative diseases, though the specific mechanisms remain unclear. This research explores the key processes behind how ferroptosis contributes to brain ageing, with a focus on the complex signaling networks that are involved. The current article aims to uncover that how ferroptosis, a specific type of cell death, may drive age-related changes in the brain. Additionally, the article also unveils its role in neurodegenerative diseases, discussing how understanding these mechanisms could open up new therapeutic avenues.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia.
| | - Saleha Anwar
- Center for Global Health Research, Saveetha medical college, Saveetha institute of Medical and Technical Sciences, Chennai, India.
| | - Kumar Venkatesan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, United Arab Emirates.
| | - Md Imtaiyaz Hassan
- Center for Interdsicplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Asimul Islam
- Center for Interdsicplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Anas Shamsi
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, United Arab Emirates.
| |
Collapse
|
10
|
Berg Y, Gabay E, Božić D, Shibli JA, Ginesin O, Asbi T, Takakura L, Mayer Y. The Impact of Nutritional Components on Periodontal Health: A Literature Review. Nutrients 2024; 16:3901. [PMID: 39599688 PMCID: PMC11597335 DOI: 10.3390/nu16223901] [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: 10/21/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease driven by the accumulation of bacterial plaque and the host's immune response, leading to the destruction of periodontal tissues. Nutrition, particularly the intake of micronutrients with anti-inflammatory and antioxidant properties, plays a crucial role in maintaining periodontal health. This review explores the impact of various micronutrients-vitamins (A, B, C, D, E), minerals (calcium, iron, zinc, potassium, copper, manganese, selenium), and omega-3 fatty acids-on periodontal disease prevention and management. Deficiencies in these nutrients can exacerbate periodontal tissue damage by impairing immune responses, promoting oxidative stress, and reducing bone and tissue regeneration capabilities. While certain populations may be more vulnerable to these deficiencies, such as those following Western diets or living in low- and middle-income countries, even in developed nations, suboptimal nutrient intake is associated with worse periodontal outcomes. Although some studies suggest that supplementation of specific micronutrients may benefit periodontal therapy, the evidence remains inconclusive, necessitating further randomized clinical trials. This review underscores the importance of considering nutritional guidance in periodontal treatment protocols and highlights the need for tailored recommendations based on recent findings.
Collapse
Affiliation(s)
- Yarden Berg
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus (RHCC), Haifa P.O. Box 9602, Israel
| | - Eran Gabay
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus (RHCC), Haifa P.O. Box 9602, Israel
- Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3101101, Israel
| | - Darko Božić
- Department of Periodontology, School of Dental Medicine, HR-10000 Zagreb, Croatia
- Department of Periodontology, University Hospital Centre Zagreb, HR-10000 Zagreb, Croatia
| | - Jamil Awad Shibli
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos 12245-000, SP, Brazil
| | - Ofir Ginesin
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus (RHCC), Haifa P.O. Box 9602, Israel
- Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3101101, Israel
| | - Thabet Asbi
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus (RHCC), Haifa P.O. Box 9602, Israel
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos 12245-000, SP, Brazil
| | - Leila Takakura
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos 12245-000, SP, Brazil
| | - Yaniv Mayer
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus (RHCC), Haifa P.O. Box 9602, Israel
- Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3101101, Israel
| |
Collapse
|
11
|
Fadón-Padilla L, Miranda-Pérez de Alejo C, Miguel-Coello AB, Beraza M, Di Silvio D, Urkola-Arsuaga A, Sánchez-Guisado MJ, Aiestaran-Zelaia I, Fernández-Méndez L, Martinez-Parra L, Ismalaj E, Berra E, Carregal-Romero S, Ruíz-Cabello J. Magnetic nanoradiotracers for targeted neutrophil detection in pulmonary arterial hypertension. J Nanobiotechnology 2024; 22:709. [PMID: 39543655 PMCID: PMC11562838 DOI: 10.1186/s12951-024-03000-7] [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: 06/27/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a severe disease characterized by elevated blood pressure in the pulmonary artery that can ultimately damage the right ventricle of the heart. PAH is pathophysiologically heterogeneous, which makes early diagnosis and treatment difficult. Inflammation is thought to be an important factor in the development and progression of this disease and may explain some of the observed interindividual differences. In the context of both acute and chronic inflammation, neutrophil recruitment to the lung has been suggested as a potential biomarker for studying PAH progression. However, there are currently no specific probes for its non-invasive in vivo detection. The imaging-based gold standard for assessing inflammation is [18F] fluorodeoxyglucose (18F-FDG), which is not cell specific. This highlights the urgent need for more specific molecular probes to support personalized medicine. METHODS This study investigated the potential of magnetic nanoradiotracers based on ultrasmall iron oxide nanoparticles, functionalized with N-cinnamoyl-F-(D)L-F-(D)L-F peptide, to detect increased neutrophil infiltration in vivo in different PAH animal models via positron emission tomography. These nanoprobes target formyl peptide receptor 1, which is abundantly expressed in the cell membrane of neutrophils. To assess the benefit of these nanoprobes, their biodistribution was first assessed via magnetic resonance imaging and histology. Then, their lung uptake was compared by positron emission tomography with that of 18F-FDG in two types of PAH animal models with different profiles of inflammation and neutrophil infiltration: monocrotaline and double-hit Sugen-chronic hypoxia PAH rat models. RESULTS Our targeted magnetic nanoradiotracer detected an increase in pulmonary neutrophil infiltration in both PAH models and distinguished between them, which was not possible with 18F-FDG PET. CONCLUSIONS This study underscores the importance of targeted imaging in providing an individualized and longitudinal evaluation of heterogeneous and multifactorial diseases such as PAH. The use of targeted multimodal nanoprobes, for magnetic resonance/positron emission tomography imaging has the potential to facilitate the diagnosis and monitoring of diseases, as well as the development of novel therapies.
Collapse
Affiliation(s)
- Lucía Fadón-Padilla
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, 21201, USA
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Claudia Miranda-Pérez de Alejo
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), Donostia, 20018, Spain
| | - Ana Beatriz Miguel-Coello
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Marta Beraza
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Desiré Di Silvio
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Ainhize Urkola-Arsuaga
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - María Jesús Sánchez-Guisado
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Irati Aiestaran-Zelaia
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), Donostia, 20018, Spain
| | - Laura Fernández-Méndez
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), Donostia, 20018, Spain
| | - Lydia Martinez-Parra
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Ermal Ismalaj
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
- Centro de investigación en red de enfermedades respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Edurne Berra
- Center for Cooperative Research in Bioscience (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, 4810, Spain
- Centro de investigación en red cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Ikerbasque, Basque Foundation for Science Ikerbasque, Bilbao, 48013, Spain
| | - Susana Carregal-Romero
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain.
- Centro de investigación en red de enfermedades respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, 28029, Spain.
- Ikerbasque, Basque Foundation for Science Ikerbasque, Bilbao, 48013, Spain.
| | - Jesús Ruíz-Cabello
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain.
- Centro de investigación en red de enfermedades respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, 28029, Spain.
- Ikerbasque, Basque Foundation for Science Ikerbasque, Bilbao, 48013, Spain.
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Madrid, 28040, Spain.
| |
Collapse
|
12
|
Tsui KH, Li CJ, Lin LT. Melatonin supplementation attenuates cuproptosis and ferroptosis in aging cumulus and granulosa cells: potential for improving IVF outcomes in advanced maternal age. Reprod Biol Endocrinol 2024; 22:138. [PMID: 39516964 PMCID: PMC11545199 DOI: 10.1186/s12958-024-01311-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Advanced maternal age is associated with decreased oocyte quantity and quality and in vitro fertilization (IVF) success rates. This study aimed to investigate whether melatonin supplementation can improve IVF outcomes in women of advanced maternal age by modulating cuproptosis and ferroptosis. METHODS This prospective cohort study included 161 women aged 35-45 years undergoing IVF-frozen embryo transfer cycles. Participants were assigned to either melatonin (n = 86, 2 mg daily for ≥ 8 weeks) or control (n = 75) groups. Cumulus cells were analyzed for cuproptosis and ferroptosis-related gene expression. Additional experiments were conducted on the HGL5 human granulosa cell line to assess mitochondrial function and metabolic reprogramming. RESULTS Melatonin supplementation significantly improved IVF outcomes in women aged ≥ 38 years, increasing clinical pregnancy rates (46.0% vs. 20.3%, P < 0.01), ongoing pregnancy rates (36.5% vs. 15.3%, P < 0.01), and live birth rates (33.3% vs. 15.3%, P < 0.05). In cumulus cells from patients, gene expression analysis revealed that melatonin modulated cuproptosis and ferroptosis-related genes, including ATP7B and GPX4, with more pronounced effects in the ≥ 38 years group. This suggests melatonin enhances cellular resilience against oxidative stress and metal-induced toxicity in the ovarian microenvironment. In vitro studies using HGL5 cells showed melatonin reduced oxidative stress markers, improved mitochondrial function, restored expression of glycolysis and TCA cycle-related genes and modulated cuproptosis and ferroptosis-related gene expression. These findings provide mechanistic insight into melatonin's protective effects against regulated cell death in ovarian cells, potentially explaining the improved IVF outcomes observed. CONCLUSIONS Melatonin supplementation significantly improved IVF outcomes in women of advanced maternal age, particularly those ≥ 38 years old, likely by modulating cuproptosis and ferroptosis and enhancing mitochondrial function in cumulus and granulosa cells. These results suggest that melatonin could be a promising adjuvant therapy for improving IVF success rates in older women.
Collapse
Affiliation(s)
- Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, 90741, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, 112, Taiwan
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, 90741, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
| | - Li-Te Lin
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan.
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, 90741, Taiwan.
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, 112, Taiwan.
| |
Collapse
|
13
|
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
|
14
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [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: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
15
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-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] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
16
|
Zavaroni A, Riva E, Borghesani V, Donati G, Santoro F, D’Amore VM, Tegoni M, Pelosi G, Buschini A, Rogolino D, Carcelli M. Synthesis and Preliminary Studies for In Vitro Biological Activity of Two New Water-Soluble Bis(thio)carbohydrazones and Their Copper(II) and Zinc(II) Complexes. Int J Mol Sci 2024; 25:10831. [PMID: 39409159 PMCID: PMC11476552 DOI: 10.3390/ijms251910831] [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: 09/11/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Research in the field of metallodrugs is continually increasing. However, it is often limited by the poor solubility in water of the metal complexes. To try to overcome this problem, the two new ligands bis-(sodium 3-methoxy-5-sulfonate-salicylaldehyde)thiocarbohydrazone (bis-TCH, Na2H4L1) and bis-(sodium 3-methoxy-5-sulfonate-salicylaldehyde)carbohydrazone (bis-CH, Na2H4L2) were synthesized and characterized, both achieving high solubility in water. The speciation of the ligands and their coordinating behaviour towards the biologically relevant Cu(II) and Zn(II) ions were studied spectroscopically and potentiometrically, determining the pKas of the ligands and the formation constants of the complex species. The monometallic and bimetallic Cu(II) and Zn(II) complexes were isolated, and the single-crystal X-ray structure of [Cu2(NaHL1)(H2O)7].3.5H2O was discussed. Finally, preliminary studies of the in vitro cytotoxic properties of the new compounds were started on normal (Hs27) and cancer (U937) cell lines. bis-TCH was able to induce a growth inhibition effect between 40% and 45% in both cell lines; bis-CH did not produce a reduction in cell viability in Hs27 cells but revealed mild antiproliferative activity after 72 h of treatment in U937 cancer cells (GI50 = 46.5 ± 4.94 μg/mL). Coordination of the Cu(II) ions increased the toxicity of the compounds, while, in contrast, Zn(II) complexes were not cytotoxic.
Collapse
Affiliation(s)
- Alessio Zavaroni
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy; (A.Z.); (E.R.); (V.B.); (M.T.); (G.P.); (A.B.); (D.R.)
| | - Elena Riva
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy; (A.Z.); (E.R.); (V.B.); (M.T.); (G.P.); (A.B.); (D.R.)
| | - Valentina Borghesani
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy; (A.Z.); (E.R.); (V.B.); (M.T.); (G.P.); (A.B.); (D.R.)
| | - Greta Donati
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (G.D.); (F.S.); (V.M.D.)
| | - Federica Santoro
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (G.D.); (F.S.); (V.M.D.)
| | - Vincenzo Maria D’Amore
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (G.D.); (F.S.); (V.M.D.)
| | - Matteo Tegoni
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy; (A.Z.); (E.R.); (V.B.); (M.T.); (G.P.); (A.B.); (D.R.)
| | - Giorgio Pelosi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy; (A.Z.); (E.R.); (V.B.); (M.T.); (G.P.); (A.B.); (D.R.)
- Centre for Molecular and Translational Oncology, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Annamaria Buschini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy; (A.Z.); (E.R.); (V.B.); (M.T.); (G.P.); (A.B.); (D.R.)
- Centre for Molecular and Translational Oncology, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Dominga Rogolino
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy; (A.Z.); (E.R.); (V.B.); (M.T.); (G.P.); (A.B.); (D.R.)
| | - Mauro Carcelli
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/A, 43124 Parma, Italy; (A.Z.); (E.R.); (V.B.); (M.T.); (G.P.); (A.B.); (D.R.)
| |
Collapse
|
17
|
Wang J, Lv C, Wei X, Li F. Molecular mechanisms and therapeutic strategies for ferroptosis and cuproptosis in ischemic stroke. Brain Behav Immun Health 2024; 40:100837. [PMID: 39228970 PMCID: PMC11369453 DOI: 10.1016/j.bbih.2024.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 09/05/2024] Open
Abstract
Ischemic stroke, as one of the most severe and prevalent neurological disorders, poses a significant threat to the health and quality of life of affected individuals. Stemming from the obstruction of blood flow, ischemic stroke, leads to cerebral tissue hypoxia and ischemia, instigating a cascade of pathophysiological changes that markedly exacerbate neuronal damage and may even culminate in cell death. In recent years, emerging research has increasingly focused on novel cell death mechanisms such as ferroptosis and cuproptosis. Mounting evidence underscores the independent roles of ferroptosis and cuproptosis in ischemic stroke. This review aims to elucidate potential cross-regulatory mechanisms between ferroptosis and cuproptosis, exploring their regulatory roles in ischemic stroke. The objective is to provide targeted therapeutic intervention strategies.
Collapse
Affiliation(s)
- Jing Wang
- Department of neurology, Lu 'an Municipal People's Hospital, Anhui, China
- Bengbu Medical College, Anhui, China
| | - Cunming Lv
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Xinyu Wei
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Feng Li
- Department of neurology, Lu 'an Municipal People's Hospital, Anhui, China
| |
Collapse
|
18
|
Song C, Wang Z, Cao J, Dong Y, Chen Y. Hesperetin alleviates aflatoxin B1 induced liver toxicity in mice: Modulating lipid peroxidation and ferritin autophagy. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116854. [PMID: 39142113 DOI: 10.1016/j.ecoenv.2024.116854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
One of the ways Aflatoxin B1 damages the liver is through ferroptosis. Ferroptosis is characterized by the build-up of lipid peroxides and reactive oxygen species (ROS) due to an excess of iron. Dietary supplements have emerged as a promising strategy for treating ferroptosis in the liver. The flavonoid component hesperetin, which is mostly present in citrus fruits, has a number of pharmacological actions, such as those against liver fibrosis, cancer, and hyperglycemia. However, hesperetin's effects and mechanisms against hepatic ferroptosis are still unknown. In this study, 24 male C57BL/6 J mice were randomly assigned to CON, AFB1 (0.45 mg/kg/day), and AFB1+ hesperetin treatment groups (40 mg/kg/day). The results showed that hesperetin improved the structural damage of the mouse liver, down-regulated inflammatory factors (Cxcl1, Cxcl2, CD80, and F4/80), and alleviated liver fibrosis induced by aflatoxin B1. Hesperetin reduced hepatic lipid peroxidation induced by iron accumulation by up-regulating the levels of antioxidant enzymes (GPX4, GSH-Px, CAT, and T-AOC). It is worth noting that hesperetin not only improved lipid peroxidation but also maintained the dynamic balance of iron ions by reducing ferritin autophagy. Mechanistically, hesperetin's ability to regulate ferritin autophagy mostly depends on the PI3K/AKT/mTOR/ULK1 pathway. In AFB1-induced HepG2 cells, the addition of PI3K inhibitor (LY294002) and AKT inhibitor (Miransertib) confirmed that hesperetin regulated the PI3K/AKT/mTOR/ULK1 pathway to inhibit ferritin autophagy and reduced the degradation of ferritin in lysosomes. In summary, our results suggest that hesperetin not only regulates the antioxidant system but also inhibits AFB1-induced ferritin hyperautophagy, thereby reducing the accumulation of iron ions to mitigate lipid peroxidation. This work provides a fresh perspective on the mechanism behind hesperetin and AFB1-induced liver damage in mice.
Collapse
Affiliation(s)
- Chao Song
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Zixu Wang
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Jing Cao
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Yulan Dong
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China.
| |
Collapse
|
19
|
Duan R, Zhao X, Hong Z, Yu L. The LINC00957/miR-17-5p axis regulates the cell cycle and migration in glioblastoma via the cuproptosis-related gene nephronectin. Transl Cancer Res 2024; 13:4923-4937. [PMID: 39430842 PMCID: PMC11483365 DOI: 10.21037/tcr-24-450] [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: 03/28/2024] [Accepted: 07/19/2024] [Indexed: 10/22/2024]
Abstract
Background Glioblastoma (GBM) is characterized by poor prognosis and a high malignancy. The competing endogenous RNA (ceRNA) network formed by long non-coding RNA (lncRNA) and microRNA (miRNA) can regulate the incidence of GBM. Therapeutic strategies targeting cuproptosis-related genes (CRGs) have helped reduce drug resistance in patients. However, the regulatory mechanism underlying the ceRNA network related to cuproptosis in GBM remains unclear. Therefore, we aim to explore the ceRNA regulatory axis associated with cuproposis in GBM and provide a new protocol for therapy. Methods The ceRNA network related to CRG was constructed by bioinformatics. Dual-luciferase reporter assay and other experiments were used to prove the conclusion. Results We found that the LINC00957/miR-17-5p axis drove nephronectin (NPNT) expression to promote the malignant progression of GBM. First, by measuring the copper ion concentration and reactive oxygen species (ROS) levels, we found that inhibiting NPNT could promote cuproptosis. Meanwhile, the results of enrichment analysis and phenotypic experiments demonstrated that the LINC00957/miR-17-5p/NPNT axis can regulate the cell cycle and migration in GBM. In terms of mechanistic evidence, findings from reporter gene experiments suggested that LINC00957 acted as a ceRNA to regulate the expression of NPNT via miR-17-5p. In addition, findings from rescue experiments confirmed that the regulation of malignant GBM progression by LINC00957 depended on NPNT to an extent. Conclusions Our findings indicate that the ceRNA regulatory network is related to cuproptosis in GBM and provide novel potential targets for the diagnosis and treatment of GBM.
Collapse
Affiliation(s)
- Renhua Duan
- Department of Neurosurgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangmao Zhao
- Department of Neurosurgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zejiang Hong
- Department of Neurosurgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lisheng Yu
- Department of Neurosurgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
20
|
Peng G, Huang Y, Xie G, Tang J. Exploring Copper's role in stroke: progress and treatment approaches. Front Pharmacol 2024; 15:1409317. [PMID: 39391696 PMCID: PMC11464477 DOI: 10.3389/fphar.2024.1409317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Copper is an important mineral, and moderate copper is required to maintain physiological processes in nervous system including cerebral ischemia/reperfusion (I/R) injury. Over the past few decades, copper induced cell death, named cuprotosis, has attracted increasing attention. Several lines of evidence have confirmed cuprotosis exerts pivotal role in diverse of pathological processes, such as cancer, neurodegenerative diseases, and I/R injury. Therefore, an in-depth understanding of the interaction mechanism between copper-mediated cell death and I/R injury may reveal the significant alterations about cellular copper-mediated homeostasis in physiological and pathophysiological conditions, as well as therapeutic strategies deciphering copper-induced cell death in cerebral I/R injury.
Collapse
Affiliation(s)
- Gang Peng
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan, China
| | - Yongpan Huang
- School of Medicine, Changsha Social Work College, Changsha, Hunan, China
| | - Guangdi Xie
- Department of Neurology, Huitong People’s Hospital, Huitong, Hunan, China
| | - Jiayu Tang
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
21
|
Zhou J, Ni W, Zhang X, Yang M, Liu X, Guo J, Li J, Zhao Q, Deng H, Lei H, Zhang L, Liao H, Jia X. Visual analysis on ferroptosis and its cross-talk to coronavirus disease 2019 (COVID-19). Heliyon 2024; 10:e37617. [PMID: 39290278 PMCID: PMC11407094 DOI: 10.1016/j.heliyon.2024.e37617] [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: 04/20/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
Background Ferroptosis is a new type of programmed cell death. Although ferroptosis has been studied in various aspects, there has been no visual analysis of ferroptosis in coronavirus disease 2019 (COVID-19) to date. It is still a global health concern of the COVID-19 pandemic worldwide, three years after its outbreak. Yet the emergence of the mutant strain Omicron has caused a fourth wave of infections in many countries. The pathogenesis of COVID-19 is still undergoing extensive exploration, which holds paramount importance in mitigating future epidemics. Methods For this study, CiteSpace 6.2 R4 software was used for bibliometric and visual atlas analysis of ferroptosis-related research, and the Genecards database was used to mine ferroptosis and COVID-19-related genes. Results We found increasing studies about ferroptosis. China and the United States have demonstrated robust scientific innovation over recent years, with extensive collaboration between their institutions and authors. Ferroptosis and COVID-19 were seen to have 13 shared genes, which may be new targets for the treatment of COVID-19 in the future. Most of the shared genes are enriched in tumor necrosis factor (TNF) pathways. The majority of those genes are up-regulated under the cellular response to oxidative stress. Genes including Tumour necrosis factor (TNF), RELA proto-oncogene (RELA), Activating transcription factor 4 (ATF4), Cytochrome b-245 beta chain (CYBB), Jun proto-oncogene (JUN), Mitogen-activated protein kinase 1 (MAPK1) and Heme oxygenase 1 (HMOX1), maybe a breakthrough for ferroptosis and COVID-19. Whilst previous research has shown there to be a relationship between ferroptosis and COVID-19, the specific role of ferroptosis remained unclear. Our study aimed to analyze the research status of ferroptosis and its relationship with COVID-19, to provide a useful reference for further prevention and treatment of COVID-19. Overall, uncovering the role of ferroptosis in SARS-CoV-2 infection is important for the development of new treatment strategies for COVID-19.
Collapse
Affiliation(s)
- Junda Zhou
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Wenjia Ni
- Key Laboratory of Non-Coding RNA and Drug Discovery at Chengdu Medical College of Sichuan Province, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Xianqin Zhang
- Key Laboratory of Non-Coding RNA and Drug Discovery at Chengdu Medical College of Sichuan Province, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Meng Yang
- Key Laboratory of Non-Coding RNA and Drug Discovery at Chengdu Medical College of Sichuan Province, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Xin Liu
- College of Public Health, Chengdu Medical College, China
| | - Jinlin Guo
- Chengdu University of Traditional Chinese Medicine, China
| | - Jian Li
- School of Basic Medical Sciences, Chengdu University, Chengdu, 610106, China
| | - Qi Zhao
- College of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Hang Deng
- Key Laboratory of Non-Coding RNA and Drug Discovery at Chengdu Medical College of Sichuan Province, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Hanyue Lei
- Key Laboratory of Non-Coding RNA and Drug Discovery at Chengdu Medical College of Sichuan Province, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Lin Zhang
- Department of Clinical Pharmacy, ShaoXing People's Hospital, ShaoXing Hospital of ZheJiang University, China
| | - Hai Liao
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xu Jia
- Key Laboratory of Non-Coding RNA and Drug Discovery at Chengdu Medical College of Sichuan Province, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| |
Collapse
|
22
|
Yu Q, Xiao Y, Guan M, Zhang X, Yu J, Han M, Li Z. Copper metabolism in osteoarthritis and its relation to oxidative stress and ferroptosis in chondrocytes. Front Mol Biosci 2024; 11:1472492. [PMID: 39329090 PMCID: PMC11425083 DOI: 10.3389/fmolb.2024.1472492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Ferroptosis, an iron-ion-dependent process of lipid peroxidation, damages the plasma membrane, leading to non-programmed cell death. Osteoarthritis (OA), a prevalent chronic degenerative joint disease among middle-aged and older adults, is characterized by chondrocyte damage or loss. Emerging evidence indicates that chondrocyte ferroptosis plays a role in OA development. However, most research has concentrated on ferroptosis regulation involving typical iron ions, potentially neglecting the significance of elevated copper ions in both serum and joint fluid of patients with OA. This review aims to fill this gap by systematically examining the interplay between copper metabolism, oxidative stress, ferroptosis, and copper-associated cell death in OA. It will provide a comprehensive overview of copper ions' role in regulating ferroptosis and their dual role in OA. This approach seeks to offer new insights for further research, prevention, and treatment of OA.
Collapse
Affiliation(s)
- Qingyuan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yanan Xiao
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mengqi Guan
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xianshuai Zhang
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jianan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mingze Han
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Zhenhua Li
- Orthopedic Center, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
23
|
Huang Q, Yang J, Zhang J, Yao L, Jiang B, Du S, Li F, Peng Q, Qin L, Wang Y, Qi L. Eupalinolide B suppresses pancreatic cancer by ROS generation and potential cuproptosis. iScience 2024; 27:110496. [PMID: 39100694 PMCID: PMC11295471 DOI: 10.1016/j.isci.2024.110496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/15/2024] [Accepted: 07/10/2024] [Indexed: 08/06/2024] Open
Abstract
Pancreatic cancer is highly lethal with limited effective treatments. This study explores the therapeutic effects of eupalinolide B (EB) from Eupatorium lindleyanum DC on pancreatic cancer cells. Through cellular functional assays, we observed that EB effectively inhibits cell viability, proliferation, migration, and invasion. In a xenograft mouse model, EB treatment resulted in reduced pancreatic cancer tumor growth and decreased expression of Ki-67. Mechanistically, EB induces apoptosis, elevates reactive oxygen species (ROS) levels, and disrupts copper homeostasis. RNA sequencing (RNA-seq) and gene set enrichment analysis (GSEA) identified copper ion binding pathways and potential involvement in cuproptosis. Furthermore, EB enhances the cytotoxic effects of elesclomol (ES), increasing ROS levels in a copper-dependent manner and exhibiting synergistic cytotoxicity. These findings suggest that EB, either alone or in combination with ES, represents a promising strategy for targeting metal ion dysregulation and inducing potential cuproptosis in pancreatic cancer, offering a potential improvement in therapeutic outcomes.
Collapse
Affiliation(s)
- Qingtian Huang
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
- Department of Pathology, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Jie Yang
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Jiaxing Zhang
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Leyi Yao
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Baoyi Jiang
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Siyuan Du
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Fengjin Li
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Qian Peng
- Biological Sample Resource Centre, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Lingsha Qin
- Biological Sample Resource Centre, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Yanfen Wang
- Department of Pathology, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| | - Ling Qi
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
- Biological Sample Resource Centre, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
- Division of Gastroenterology, Institute of Digestive Disease, the Affiliated Qingyuan Hospital (Qingyuan Peoples's Hospital), Guangzhou Medical University, Qingyuan 511518, Guang Dong, China
| |
Collapse
|
24
|
Tang Y, Liang H, Su L, Xue X, Zhan J. Ferroptosis: a new perspective on the pathogenesis of radiation-induced cataracts. Front Public Health 2024; 12:1449216. [PMID: 39220446 PMCID: PMC11363423 DOI: 10.3389/fpubh.2024.1449216] [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: 06/14/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Ionizing radiation is a significant risk factor for cataracts, but the pathogenesis of radiation-induced cataracts remains incompletely understood. Ferroptosis, an iron-dependent form of programmed cell death discovered in recent years, has gained increasing attention for its role in various diseases. This article systematically reviews research progress on ionizing radiation, ferroptosis, age-related cataracts, and radiation-induced cataracts. It proposes the "ferroptosis hypothesis" for the pathogenesis of radiation-induced cataracts. Through ionization and oxidative stress effects, ionizing radiation leads to elevated free iron levels and exacerbated lipid peroxidation in lens cells, activating the ferroptosis pathway and resulting in lens opacity. The involvement of ferroptosis in the development of age-related cataracts suggests that it may also be an important pathogenic mechanism of radiation-induced cataracts. Targeting the ferroptosis pathway may be a novel strategy for preventing and treating radiation-induced cataracts. Furthermore, developing new ferroptosis-specific inhibitors with improved targeting and pharmacokinetic properties is also an essential direction for research on preventing and treating radiation-induced cataracts. The study of ferroptosis provides new insights into the mechanism and management of radiation-induced cataracts, potentially transforming radiation-induced cataracts from "inevitable" to "preventable and treatable."
Collapse
Affiliation(s)
| | | | | | - Xiangming Xue
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan, China
| | - Jingming Zhan
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan, China
| |
Collapse
|
25
|
Fujii J, Imai H. Oxidative Metabolism as a Cause of Lipid Peroxidation in the Execution of Ferroptosis. Int J Mol Sci 2024; 25:7544. [PMID: 39062787 PMCID: PMC11276677 DOI: 10.3390/ijms25147544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Ferroptosis is a type of nonapoptotic cell death that is characteristically caused by phospholipid peroxidation promoted by radical reactions involving iron. Researchers have identified many of the protein factors that are encoded by genes that promote ferroptosis. Glutathione peroxidase 4 (GPX4) is a key enzyme that protects phospholipids from peroxidation and suppresses ferroptosis in a glutathione-dependent manner. Thus, the dysregulation of genes involved in cysteine and/or glutathione metabolism is closely associated with ferroptosis. From the perspective of cell dynamics, actively proliferating cells are more prone to ferroptosis than quiescent cells, which suggests that radical species generated during oxygen-involved metabolism are responsible for lipid peroxidation. Herein, we discuss the initial events involved in ferroptosis that dominantly occur in the process of energy metabolism, in association with cysteine deficiency. Accordingly, dysregulation of the tricarboxylic acid cycle coupled with the respiratory chain in mitochondria are the main subjects here, and this suggests that mitochondria are the likely source of both radical electrons and free iron. Since not only carbohydrates, but also amino acids, especially glutamate, are major substrates for central metabolism, dealing with nitrogen derived from amino groups also contributes to lipid peroxidation and is a subject of this discussion.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Hirotaka Imai
- Laboratory of Hygienic Chemistry, School of Pharmaceutical Sciences, Kitasato University, Tokyo 108-8641, Japan
- Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo 108-8641, Japan
| |
Collapse
|
26
|
Liu S, Wei Y, Liang Y, Du P, Lei P, Yu D, Zhang H. Engineering Nanozymes for Tumor Therapy via Ferroptosis Self-Amplification. Adv Healthc Mater 2024; 13:e2400307. [PMID: 38573778 DOI: 10.1002/adhm.202400307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Ferroptosis induction is an emerging strategy for tumor therapy. Reactive oxygen species (ROS) can induce ferroptosis but are easily consumed by overexpressed glutathione (GSH) in tumor cells. Therefore, achieving a large amount of ROS production in tumor cells without being consumed is key to efficiently inducing ferroptosis. In this study, a self-amplifying ferroptosis-inducing therapeutic agent, Pd@CeO2-Fe-Co-WZB117-DSPE-PEG-FA (PCDWD), is designed for tumor therapy. PCDWD exhibits excellent multi-enzyme activities due to the loading of Fe-Co dual atoms with abundant active sites, including peroxidase-like enzymes, catalase-like enzymes, and glutathione oxidases (GSHOx), which undergo catalytic reactions in the tumor microenvironment to produce ROS, thereby inducing ferroptosis. Furthermore, PCDWD can also deplete GSH in tumor cells, thus reducing the consumption of ROS by GSH and inhibiting the expression of GSH peroxidase 4. Moreover, the photothermal effect of PCDWD can not only directly kill tumor cells but also further enhance its own enzyme activities, consequently promoting ferroptosis in tumor cells. In addition, WZB117 can reduce the expression of heat shock protein 90 by inhibiting glucose transport, thereby reducing the thermal resistance of tumor cells and further improving the therapeutic effect. Finally, X-ray computed tomography imaging of PCDWD guides it to achieve efficient tumor therapy.
Collapse
Affiliation(s)
- Shuyu Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yi Wei
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Yuan Liang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Pengye Du
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Pengpeng Lei
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Duo Yu
- Second Hospital of Jilin University, The Second Hospital of Jilin University Department of Radiotherapy, Changchun, 130062, China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
27
|
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 2024:10.1007/s11010-024-05056-3. [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] [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
|
28
|
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
|
29
|
Teschke R, Eickhoff A. Wilson Disease: Copper-Mediated Cuproptosis, Iron-Related Ferroptosis, and Clinical Highlights, with Comprehensive and Critical Analysis Update. Int J Mol Sci 2024; 25:4753. [PMID: 38731973 PMCID: PMC11084815 DOI: 10.3390/ijms25094753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Wilson disease is a genetic disorder of the liver characterized by excess accumulation of copper, which is found ubiquitously on earth and normally enters the human body in small amounts via the food chain. Many interesting disease details were published on the mechanistic steps, such as the generation of reactive oxygen species (ROS) and cuproptosis causing a copper dependent cell death. In the liver of patients with Wilson disease, also, increased iron deposits were found that may lead to iron-related ferroptosis responsible for phospholipid peroxidation within membranes of subcellular organelles. All topics are covered in this review article, in addition to the diagnostic and therapeutic issues of Wilson disease. Excess Cu2+ primarily leads to the generation of reactive oxygen species (ROS), as evidenced by early experimental studies exemplified with the detection of hydroxyl radical formation using the electron spin resonance (ESR) spin-trapping method. The generation of ROS products follows the principles of the Haber-Weiss reaction and the subsequent Fenton reaction leading to copper-related cuproptosis, and is thereby closely connected with ROS. Copper accumulation in the liver is due to impaired biliary excretion of copper caused by the inheritable malfunctioning or missing ATP7B protein. As a result, disturbed cellular homeostasis of copper prevails within the liver. Released from the liver cells due to limited storage capacity, the toxic copper enters the circulation and arrives at other organs, causing local accumulation and cell injury. This explains why copper injures not only the liver, but also the brain, kidneys, eyes, heart, muscles, and bones, explaining the multifaceted clinical features of Wilson disease. Among these are depression, psychosis, dysarthria, ataxia, writing problems, dysphagia, renal tubular dysfunction, Kayser-Fleischer corneal rings, cardiomyopathy, cardiac arrhythmias, rhabdomyolysis, osteoporosis, osteomalacia, arthritis, and arthralgia. In addition, Coombs-negative hemolytic anemia is a key feature of Wilson disease with undetectable serum haptoglobin. The modified Leipzig Scoring System helps diagnose Wilson disease. Patients with Wilson disease are well-treated first-line with copper chelators like D-penicillamine that facilitate the removal of circulating copper bound to albumin and increase in urinary copper excretion. Early chelation therapy improves prognosis. Liver transplantation is an option viewed as ultima ratio in end-stage liver disease with untreatable complications or acute liver failure. Liver transplantation finally may thus be a life-saving approach and curative treatment of the disease by replacing the hepatic gene mutation. In conclusion, Wilson disease is a multifaceted genetic disease representing a molecular and clinical challenge.
Collapse
Affiliation(s)
- Rolf Teschke
- Department of Internal Medicine II, Division of Gastroenterology and Hepatology, Klinikum Hanau, D-63450 Hanau, Germany;
- Academic Teaching Hospital of the Medical Faculty, Goethe University Frankfurt, D-60590 Frankfurt, Germany
| | - Axel Eickhoff
- Department of Internal Medicine II, Division of Gastroenterology and Hepatology, Klinikum Hanau, D-63450 Hanau, Germany;
- Academic Teaching Hospital of the Medical Faculty, Goethe University Frankfurt, D-60590 Frankfurt, Germany
| |
Collapse
|
30
|
Chen Z, Du D, Li J, Zhang W, Shao J. Cuproptosis-related molecular classification and gene signature of hepatocellular carcinoma and experimental verification. Transl Cancer Res 2024; 13:1268-1289. [PMID: 38617510 PMCID: PMC11009816 DOI: 10.21037/tcr-23-1876] [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: 10/10/2023] [Accepted: 02/08/2024] [Indexed: 04/16/2024]
Abstract
Background Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy with poor overall prognosis. Cuproptosis, a recently proposed mode of copper-dependent cell death, plays a critical role in the malignant progression of various tumors; however, the expression and prognostic value of cuproptosis-related regulatory genes in HCC remain unclear. Methods Genomic, genetic, and expression profiles of ten key cuproptosis-related regulatory genes were analyzed using The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) dataset and protein expression data from the Human Protein Atlas (HPA) database. Unsupervised clustering of HCC patients based on these ten key cuproptosis-related regulatory genes was used to identify different HCC subtypes and analyze the differences in clinical and immune characteristics among subtypes. Subsequently, univariate Cox and least absolute shrinkage and selection operator (LASSO) Cox analyses were used to establish a cuproptosis-related prognostic signature, and the accuracy of prognostic signature prediction was internally validated by Kaplan-Meier survival analysis and time-dependent receiver operating characteristic curve in TCGA training and testing cohorts. The prognostic signature was externally validated using TCGA-LIHC entire cohort and International Cancer Genome Consortium Liver Cancer (ICGC-LIRI) cohorts. Finally, the expression landscape of cuproptosis-related regulatory genes in prognostic signature was explored by quantitative real-time polymerase chain reaction (qRT-PCR), western blotting and immunohistochemistry (IHC) experiments. Results Ten cuproptosis-related genes were differentially expressed in normal and HCC tissues. Unsupervised clustering identified two subtypes and HCC patients with these two subtypes had different clinical prognoses and immune characteristics, as well as different degrees of response to immunotherapy. Lipoyltransferase 1 (LIPT1), dihydrolipoamide s-acetyltransferase (DLAT), and cyclin dependent kinase inhibitor 2A (CDKN2A) were selected to construct a prognostic signature, which significantly distinguished HCC patients with different survival periods in the TCGA training and testing cohorts and was well validated in both the TCGA-LIHC entire cohort and ICGC-LIRI cohort. The risk score of the prognostic signature was confirmed to be an independent prognostic factor, and nomograms were generated to effectively predict the probability of HCC patient survival. The qRT-PCR, western blotting and IHC results also revealed a significant imbalance in the expression of these cuproptosis-related genes in HCC. Conclusions The classification and prognostic signature based on cuproptosis-related regulatory genes helps to explain the heterogeneity of HCC, which may contribute to the individualized treatment of patients with the disease.
Collapse
Affiliation(s)
- Zehao Chen
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Engineering Research Center of Hepatobiliary Disease, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dongnian Du
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Engineering Research Center of Hepatobiliary Disease, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiajuan Li
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Engineering Research Center of Hepatobiliary Disease, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenming Zhang
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Engineering Research Center of Hepatobiliary Disease, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianghua Shao
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Engineering Research Center of Hepatobiliary Disease, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
31
|
Demir E, Turna Demir F. Genotoxicity responses of single and mixed exposure to heavy metals (cadmium, silver, and copper) as environmental pollutants in Drosophila melanogaster. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 106:104390. [PMID: 38367919 DOI: 10.1016/j.etap.2024.104390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
Heavy metals are now persistently present in living things' environments, in addition to their potential toxicity. Therefore, the aim of this study was to utilize D. melanogaster to determine the biological effects induced by different heavy metals including cadmium chloride (CdCl2), copper (II) sulfate pentahydrate (CuSO 4.5 H2O), and silver nitrate (AgNO3). In vivo experiments were conducted utilizing three low and environmentally relevant concentrations from 0.01 to 0.5 mM under single and combined exposure scenarios on D. melanogaster larvae. The endpoints measured included viability, reactive oxygen species (ROS) generation and genotoxic effects using Comet assay and the wing-spot test. Results indicated that tested heavy metals were not toxic in the egg-to adult viability. However, combined exposure (CdCl2+AgNO3 and CdCl2+AgNO3+CuSO 4.5 H2O) resulted in significant genotoxic and unfavorable consequences, as well as antagonistic and/or synergistic effects on oxidative damage and genetic damage.
Collapse
Affiliation(s)
- Eşref Demir
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Services and Techniques, Medical Laboratory Techniques Programme, Vocational School of Health Services, Antalya Bilim University, Dosemealti, Antalya 07190, Turkey.
| | - Fatma Turna Demir
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; Department of Medical Services and Techniques, Medical Laboratory Techniques Programme, Vocational School of Health Services, Antalya Bilim University, Dosemealti, Antalya 07190, Turkey
| |
Collapse
|
32
|
Wang J, Li J, Liu J, Chan KY, Lee HS, Lin KN, Wang CC, Lau TS. Interplay of Ferroptosis and Cuproptosis in Cancer: Dissecting Metal-Driven Mechanisms for Therapeutic Potentials. Cancers (Basel) 2024; 16:512. [PMID: 38339263 PMCID: PMC10854932 DOI: 10.3390/cancers16030512] [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: 12/06/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Iron (Fe) and copper (Cu), essential transition metals, play pivotal roles in various cellular processes critical to cancer biology, including cell proliferation, mitochondrial respiration, distant metastases, and oxidative stress. The emergence of ferroptosis and cuproptosis as distinct forms of non-apoptotic cell death has heightened their significance, particularly in connection with these metal ions. While initially studied separately, recent evidence underscores the interdependence of ferroptosis and cuproptosis. Studies reveal a link between mitochondrial copper accumulation and ferroptosis induction. This interconnected relationship presents a promising strategy, especially for addressing refractory cancers marked by drug tolerance. Harnessing the toxicity of iron and copper in clinical settings becomes crucial. Simultaneous targeting of ferroptosis and cuproptosis, exemplified by the combination of sorafenib and elesclomol-Cu, represents an intriguing approach. Strategies targeting mitochondria further enhance the precision of these approaches, providing hope for improving treatment outcomes of drug-resistant cancers. Moreover, the combination of iron chelators and copper-lowering agents with established therapeutic modalities exhibits a synergy that holds promise for the augmentation of anti-tumor efficacy in various malignancies. This review elaborates on the complex interplay between ferroptosis and cuproptosis, including their underlying mechanisms, and explores their potential as druggable targets in both cancer research and clinical settings.
Collapse
Affiliation(s)
- Jinjiang Wang
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Jiaxi Li
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jiao Liu
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Kit-Ying Chan
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Ho-Sze Lee
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Kenneth Nansheng Lin
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Chi-Chiu Wang
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Tat-San Lau
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| |
Collapse
|