1
|
Liao C, Qin Z, Xu J, Lan T, Shi S, Guo Y. MnO 2 nanozyme-based dual-mode colorimetric and fluorescence determination of antioxidant activity and HPLC - UV - MS/MS profiling of antioxidants. Mikrochim Acta 2025; 192:386. [PMID: 40439768 DOI: 10.1007/s00604-025-07158-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 04/06/2025] [Indexed: 06/11/2025]
Abstract
MnO2 nanozyme-based strategy is for the first time exploited for dual-mode colorimetric and fluorescence determination of antioxidant activity and HPLC - UV - MS/MS profiling of antioxidants. MnO2 nanosheets (MnO2 NS) with outstanding oxidase-like property can trigger rhodamine B (RhB) chromogenic reaction, leading to design an accurate, selective, and sensitive dual-mode colorimetric and fluorescence method for total antioxidant capacity (TAC) determination. Additionally, antioxidants in complex extract can react with MnO2 NS catalytic reactive oxygen species (ROS) intermediates (•O2‒ and 1O2), which deduced the HPLC peaks decreased or disappeared. And screened antioxidants can be identified by MS/MS analysis. As proof of concept, antioxidant levels of four flavonoids (quercetin, rutin, hesperidin, and nobiletin) with different substituent groups and six samples (peels, pulps, and juices for two citrus cultivars ChunJian tangerine and Debao navel orange) were successfully measured. Nineteen flavonoids with ROS scavenging activity from citrus samples have been screened out and characterized. Especially, polymethoxyflavones, nonactive in DPPH•/ABTS•+-based assays, presented certain ROS scavenging activity. Together, this study provided a novel and efficient platform to accurately and selectively measure and screen physiological antioxidants in real complex samples, revealing its great promise and convenient applications in future.
Collapse
Affiliation(s)
- Chunhui Liao
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan, China
| | - Ziyi Qin
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan, China
| | - Jinju Xu
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan, China
| | - Tian Lan
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan, China
| | - Shuyun Shi
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan, China.
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, Hunan, China.
| | - Ying Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
2
|
Chang L, Qin C, Wu J, Jiang H, Xu Q, Chen J, Xu X, Zhang X, Guan M, Deng X. The crosstalk between glutathione metabolism and non-coding RNAs in cancer progression and treatment resistance. Redox Biol 2025; 84:103689. [PMID: 40403492 DOI: 10.1016/j.redox.2025.103689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 05/11/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025] Open
Abstract
Excessive reactive oxygen species (ROS) are closely associated with the initiation and progression of cancers. As the most abundant intracellular antioxidant, glutathione (GSH) plays a critical role in regulating cellular ROS levels, modulating physiological processes, and is intricately linked to tumor progression and drug resistance. However, the underlying mechanisms remain not fully elucidated. Non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), are key regulators of GSH levels. Different ncRNAs modulate various pathways involved in GSH metabolism, and these regulatory targets have the potential to serve as therapeutic targets for enhancing cancer treatment. In this review, we summarize the functions of GSH metabolism and highlight the significance of ncRNA-mediated regulation of GSH in cancer progression, drug resistance, and clinical applications.
Collapse
Affiliation(s)
- Lu Chang
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Chao Qin
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Jianbo Wu
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Haoqin Jiang
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Qianqian Xu
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Jian Chen
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Xiao Xu
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Xinju Zhang
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China
| | - Ming Guan
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China.
| | - Xuan Deng
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai, 200040, China.
| |
Collapse
|
3
|
Ma X, Zhang B, Yin X, Yang S, Lin Z, Yang Y, Zhou X. CPT1A/HIF-1α positive feedback loop induced fatty acid oxidation metabolic pathway contributes to the L-ascorbic acid-driven angiogenesis in breast cancer. Breast Cancer Res 2025; 27:74. [PMID: 40355947 PMCID: PMC12067761 DOI: 10.1186/s13058-025-02039-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND In tumors rich in adipose tissue, angiogenesis is a critical factor in promoting cancer cell metastasis. However, the connection between angiogenesis and the mechanisms driving adipose metabolic remodeling in breast cancer (BC) remains insufficiently understood. This research seeks to explore whether and how CPT1A, a crucial rate-limiting enzyme in fatty acid oxidation (FAO), supports angiogenesis through metabolic pathways in BC. METHODS First, cell functional assays and animal models were employed to elucidate the pro-carcinogenic effects of CPT1A on BC and its role in metabolic alterations. Following this, the reciprocal regulatory relationship between CPT1A and HIF-1α was elucidated using transcriptomic studies, ubiquitination analysis, and dual-luciferase assays. Matrigel tube formation assays, vasculogenic mimicry assays, and chick chorioallantoic membrane (CAM) assays were utilized to evaluate the effect of CPT1A on the pro-angiogenic properties of BC. Subsequently, untargeted metabolomics was employed to identify specific metabolic changes in supernatants with and without CPT1A expression and verified by functional recovery experiments. Finally, the prognostic significance of CPT1A and the vascular marker VEGF in BC tissues was evaluated using tissue microarrays and public databases. RESULTS CPT1A overexpression significantly enhanced cell proliferation, motility, and angiogenesis via activating the FAO metabolic pathway, as demonstrated by both in vivo and in vitro experiments. Mechanistically, CPT1A regulates the ubiquitination level of hypoxia-inducible factor-1α (HIF-1α), which directly binds to the CPT1A promoter. Mutations at the 63-74 and 434-445 regions significantly reduced CPT1A promoter activity, indicating that these sites are critical for its transcriptional regulation. Ultimately, this interaction creates a reinforcing feedback loop between CPT1A and HIF-1α. Subsequently, this feedback loop alters changes in extracellular L-ascorbic acid (LAA) levels. Interestingly, LAA affects ROS homeostasis through the Nrf2/NQO1 pathway, specifically influencing angiogenesis in BC and HUVECs, while having no significant effect on their proliferation or EMT process. Moreover, increased expression levels of CPT1A and vascular endothelial growth factor (VEGF) were significantly associated with lymph node metastasis and adverse outcomes in BC patients. CONCLUSION The CPT1A/HIF-1α positive feedback loop critically regulates angiogenesis through activation of the Nrf2/NQO1 pathway, modulated by LAA. These findings highlight CPT1A and VEGF as promising therapeutic targets and prognostic biomarkers for angiogenesis in BC.
Collapse
Affiliation(s)
- Xiao Ma
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, PR China
- Key Laboratory of Tumor Pathobiology, Yanbian University, State Ethnic Affairs Commission, Yanji, 133000, PR China
| | - Baojian Zhang
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, PR China
| | - Xuezhe Yin
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, PR China
| | - ShiPeng Yang
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, PR China
| | - Zhenhua Lin
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, PR China
- Key Laboratory of Tumor Pathobiology, Yanbian University, State Ethnic Affairs Commission, Yanji, 133000, PR China
| | - Yang Yang
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, PR China.
- Key Laboratory of Tumor Pathobiology, Yanbian University, State Ethnic Affairs Commission, Yanji, 133000, PR China.
| | - Xianchun Zhou
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, PR China.
| |
Collapse
|
4
|
Shi Y, Wang J, Zhang Y, Wu K, Zhu Y, Yan K, Ouyang Q. Effectiveness of herbal medicine for liver cancer treatment as revealed by a bibliometric and visualization analysis. Front Oncol 2025; 15:1527091. [PMID: 40406241 PMCID: PMC12094935 DOI: 10.3389/fonc.2025.1527091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Background Liver cancer is highly prevalent worldwide. However, current medical treatments remain insufficient. Although herbal medicine has a long history and extensive expertise in treating liver cancer, the literature in this field has not been thoroughly explored. This study aims to assess and analyze the distribution patterns and key research areas of publications concerning herbal medicine for liver cancer. Methods Literature on herbal medicine and liver cancer published between January 1, 2008, and September 28, 2024, was collected for this research. Excel, CiteSpace 6.4.R1, VOSviewer 1.6.20, Scimago Graphica, and Bibliometrix 4.1 were used for data analysis. Result The study examines 634 academic articles on herbal medicine for liver cancer, with the majority contributed by Chinese researchers, particularly from Shanghai University of Traditional Chinese Medicine. Wang Ning is the most productive author, possessing the highest h-index. The JOURNAL OF ETHNOPHARMACOLOGY has the most publications and the highest h-index. Journals publishing on herbal medicine and liver cancer are primarily in the fields of molecular biology and immunology, whereas the cited journals are mainly in the fields of environment, toxicology, and nutrition. Keyword clustering analysis indicates that "NF kappa B" and apoptosis have long been the main research topics in this field. Analysis of emergent words suggests that "network pharmacology", antioxidants, "adjuvant therapy", and "molecular docking" may become significant research topics in the near future. Conclusion This analysis provides a comprehensive overview of the current status, primary focuses, and emerging trends in research related to herbal medicine and liver cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qin Ouyang
- Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| |
Collapse
|
5
|
An Y, Liu W, Deng Y, Huang W, Huang J. SLC7A11-HSPB1 Axis: A Novel Mechanism for Hepatocellular Carcinoma Progression and Ferroptosis Regulation. Biomed J 2025:100869. [PMID: 40339903 DOI: 10.1016/j.bj.2025.100869] [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: 11/19/2024] [Revised: 03/27/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND SLC7A11, a plasma membrane protein, has been implicated as an oncogene in various cancers, including hepatocellular carcinoma (HCC). Its role in HCC pathogenesis, particularly in relation to ferroptosis, is not well understood. This study aims to investigate the function of SLC7A11 with ferroptosis and its interaction in development of HCC. METHODS AND MATERIAL Clinical HCC tissue samples were used to analyze the expression of SLC7A11 by RT-PCR. The impact of SLC7A11 on HCC cell viability, proliferation, and migration was assessed by CCK-8, AlamarBlue and Transwell. Protein-protein interactions were explored using co-immunoprecipitation and immunofluorescence. The effect of SLC7A11 on ferroptosis was evaluated by iron levels, ROS, and GSH. The impact of sorafenib and doxorubicin (DOX) on HCC cells was analyzed using cell viability assay. RESULTS SLC7A11 was found to be highly expressed in HCC tissues and was correlated with tumor size and poor prognosis. Overexpression of SLC7A11 in HCC cells promoted cell viability, proliferation, and migration. Additionally, SLC7A11 overexpression mitigated erastin-induced ferroptosis, as evidenced by decreased ROS levels and increased GSH levels. We also discovered that SLC7A11 interacted with HSPB1. HSPB1 inhibited erastin-induced ferroptosis. Furthermore, a portion of the cell death induced by sorafenib and DOX is attributed to ferroptosis, with HSPB1 and SLC7A11 inhibiting the death induced by the two drugs, respectively. CONCLUSIONS SLC7A11 plays a significant role in HCC progression by inhibiting ferroptosis, and its interaction with HSPB1 is a critical pathway in this process. Targeting the SLC7A11-HSPB1 axis may provide a novel therapeutic strategy for HCC treatment, highlighting the importance of understanding the mechanisms of ferroptosis in cancer cells.
Collapse
Affiliation(s)
- Yan An
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China; Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200050, China
| | - Weilong Liu
- Institute of Hepatology, National clinical research center for infectious diseases, Guangdong Key Lab for Diagnosis &Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
6
|
Sulaiman C, George BP, Balachandran I, Abrahamse H. Cancer and Traditional Medicine: An Integrative Approach. Pharmaceuticals (Basel) 2025; 18:644. [PMID: 40430464 PMCID: PMC12114720 DOI: 10.3390/ph18050644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Despite numerous advances in treatment, cancer still remains a leading cause of death worldwide. Given the significant health and economic burden this disease imposes, it is important to explore more effective treatment strategies. A major drawback of conventional cancer therapies is the persistence of drug resistance, adverse reactions to chemotherapy, digestive damage, reduced quality of life, and high treatment costs. To address these challenges, researchers have been investigating the utility of using complementary and alternative medicine (CAM) alongside conventional cancer treatments. Some of these CAM approaches have been reported to enhance patients' quality of life and reduce the severity of adverse effects from conventional therapies. This review explores the utility of traditional Chinese, Korean, Indian, Japanese, and South African medicines as supportive therapies for conventional cancer treatments. We also discuss the concept of integrative oncology and its global relevance, as CAM approaches typically emphasize whole-person care, including diet, lifestyle, and mental/emotional well-being. In addition, we identify key active phytochemicals and herbal medicines used in these traditional systems for cancer treatment. Our discussion aims to provide a foundation for future research into integrative oncology, fostering an interdisciplinary and complementary approach to cancer management.
Collapse
Affiliation(s)
- Cheruthazhakkat Sulaiman
- Phytochemistry Division, Centre for Medicinal Plants Research, Arya Vaidya Sala, Kottakkal 676503, Kerala, India; (C.S.); (I.B.)
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| | - Blassan P. George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| | - Indira Balachandran
- Phytochemistry Division, Centre for Medicinal Plants Research, Arya Vaidya Sala, Kottakkal 676503, Kerala, India; (C.S.); (I.B.)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| |
Collapse
|
7
|
Gujar V, Li H, Paull TT, Neumann CA, Weyemi U. Unraveling the nexus: Genomic instability and metabolism in cancer. Cell Rep 2025; 44:115540. [PMID: 40208791 PMCID: PMC12043202 DOI: 10.1016/j.celrep.2025.115540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025] Open
Abstract
The DNA-damage response (DDR) is a signaling network that enables cells to detect and repair genomic damage. Over the past three decades, inhibiting DDR has proven to be an effective cancer therapeutic strategy. Although cancer drugs targeting DDR have received approval for treating various cancers, tumor cells often develop resistance to these therapies, owing to their ability to undergo energetic metabolic reprogramming. Metabolic intermediates also influence tumor cells' ability to sense oxidative stress, leading to impaired redox metabolism, thus creating redox vulnerabilities. In this review, we summarize recent advances in understanding the crosstalk between DDR and metabolism. We discuss combination therapies that target DDR, metabolism, and redox vulnerabilities in cancer. We also outline potential obstacles in targeting metabolism and propose strategies to overcome these challenges.
Collapse
Affiliation(s)
- Vaibhavi Gujar
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haojian Li
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tanya T Paull
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Carola A Neumann
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Urbain Weyemi
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
8
|
Kharchenko O, Gouju J, Verdu I, Bastiat G, Hudhomme P, Passirani C, Saulnier P, Krupka O. Heavy-Atom-Free Photosensitizer-Loaded Lipid Nanocapsules for Photodynamic Therapy. ACS APPLIED BIO MATERIALS 2025; 8:3086-3095. [PMID: 40113596 DOI: 10.1021/acsabm.4c01953] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Photodynamic therapy (PDT) is a clinically approved noninvasive treatment for cancer that employs a photosensitizer (PS) to generate cytotoxic reactive singlet oxygen (ROS) species that precisely destroy cancer cells at the targeted tumor sites. There is growing interest in the development of innovative photosensitizing agents and advanced delivery methods, offering superior phototherapeutic performance. The delivery of PS is a challenging task in PDT in regard to the high hydrophobicity of the PS molecule. To address this challenge, the incorporation of heavy-atom-free PS (HAF-PS) in effective drug delivery carriers is promising for PDT improvement. Herein, we propose a strategy to encapsulate the HAF-PS from the perylenediimide (PDI) family in the oily core of lipid nanocapsules (LNCs). The resulting HAF-PS-loaded LNCs formulations have the advantage to efficiently generate singlet oxygen (1O2) in a biorelevant environment. The LNCs formulations loaded with O-PDI (O-PDI@LNC) and 1S-PDI (1S-PDI@LNC) were obtained by a solvent-free phase-inversion temperature (PIT) method. Our study demonstrates that optimized LNCs formulation loaded with 1S-PDI acting as PS is a highly efficient approach to deliver phototherapeutic agents for PDT. Overall, it has been shown that illumination of 1S-PDI leads to dramatic 1O2 production with an impressive quantum yield (φSOQY = 0.94) which was tested with 1,3-diphenylisobenzofuran (DPBF) as a specific trap. Moreover, the 1O2 generation was calculated in a phosphate buffer solution (φSOQY = 0.52) for loaded nanocarrier 1S-PDI@LNC. In vitro cytotoxicity studies demonstrated a low dark toxicity of 1S-PDI@LNC while illumination significantly enhanced its photocytotoxicity in cells. Furthermore, the cellular internalization of LNCs was demonstrated in U-87 MG cells using O-PDI@LNC as a model, exploiting the excellent fluorescence properties of O-PDI. This study has significant potential for advancing the development of HAF-PS-loaded LNCs for minimally invasive PDT.
Collapse
Affiliation(s)
| | - Julien Gouju
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
- Laboratory of Neurobiology and Neuropathology, University-Hospital of Angers, F-49933 Angers, France
| | - Isabelle Verdu
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | | | - Piétrick Hudhomme
- Univ Angers, CNRS, MOLTECH-Anjou, SFR MATRIX, F-49000 Angers, France
| | | | - Patrick Saulnier
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Oksana Krupka
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| |
Collapse
|
9
|
Wang X, Xue Y, Chang L, Zhu X, Liu W, Liang T. The Regulation of Trace Metal Elements in Cancer Ferroptosis. Adv Biol (Weinh) 2025:e2400821. [PMID: 40200790 DOI: 10.1002/adbi.202400821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/11/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, as novel type of regulated cell death that has garnered widespread attention over the past decade, has witnessed the continuous discovery of an increasing number of regulatory mechanisms. Trace metal elements play a multifaceted and crucial role in oncology. Interestingly, it has been increasingly evident that these elements, such as copper, are involved in the regulation of iron accumulation, lipid peroxidation and antiferroptotic systems, suggesting the existence of "nonferrous" mechanisms in ferroptosis. In this review, a comprehensive overview of the composition and mechanism of ferroptosis is provided. The interaction between copper metabolism (including cuproptosis) and ferroptosis in cancer, as well as the roles of other trace metal elements (such as zinc, manganese, cobalt, and molybdenum) in ferroptosis are specifically focused. Furthermore, the applications of nanomaterials based on these metals in cancer therapy are also reviewed and potential strategies for co-targeting ferroptosis and cuproptosis are explored. Nevertheless, in light of the intricate and ambiguous nature of these interactions, ongoing research is essential to further elucidate the "nonferrous" mechanisms of ferroptosis, thereby facilitating the development of novel therapeutic targets and approaches for cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuanyuan Xue
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lei Chang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xuena Zhu
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Wenjun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
- Zhejiang University Cancer Center, Hangzhou, 310003, China
| |
Collapse
|
10
|
Li B, Ming H, Qin S, Nice EC, Dong J, Du Z, Huang C. Redox regulation: mechanisms, biology and therapeutic targets in diseases. Signal Transduct Target Ther 2025; 10:72. [PMID: 40050273 PMCID: PMC11885647 DOI: 10.1038/s41392-024-02095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/09/2024] [Accepted: 11/21/2024] [Indexed: 03/09/2025] Open
Abstract
Redox signaling acts as a critical mediator in the dynamic interactions between organisms and their external environment, profoundly influencing both the onset and progression of various diseases. Under physiological conditions, oxidative free radicals generated by the mitochondrial oxidative respiratory chain, endoplasmic reticulum, and NADPH oxidases can be effectively neutralized by NRF2-mediated antioxidant responses. These responses elevate the synthesis of superoxide dismutase (SOD), catalase, as well as key molecules like nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH), thereby maintaining cellular redox homeostasis. Disruption of this finely tuned equilibrium is closely linked to the pathogenesis of a wide range of diseases. Recent advances have broadened our understanding of the molecular mechanisms underpinning this dysregulation, highlighting the pivotal roles of genomic instability, epigenetic modifications, protein degradation, and metabolic reprogramming. These findings provide a foundation for exploring redox regulation as a mechanistic basis for improving therapeutic strategies. While antioxidant-based therapies have shown early promise in conditions where oxidative stress plays a primary pathological role, their efficacy in diseases characterized by complex, multifactorial etiologies remains controversial. A deeper, context-specific understanding of redox signaling, particularly the roles of redox-sensitive proteins, is critical for designing targeted therapies aimed at re-establishing redox balance. Emerging small molecule inhibitors that target specific cysteine residues in redox-sensitive proteins have demonstrated promising preclinical outcomes, setting the stage for forthcoming clinical trials. In this review, we summarize our current understanding of the intricate relationship between oxidative stress and disease pathogenesis and also discuss how these insights can be leveraged to optimize therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Bowen Li
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Hui Ming
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Siyuan Qin
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jingsi Dong
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhongyan Du
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China.
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China.
| |
Collapse
|
11
|
Katopodi V, Marino A, Pateraki N, Verheyden Y, Cinque S, Jimenez EL, Adnane S, Demesmaeker E, Scomparin A, Derua R, Groaz E, Leucci E. The long non-coding RNA ROSALIND protects the mitochondrial translational machinery from oxidative damage. Cell Death Differ 2025; 32:397-415. [PMID: 39294440 PMCID: PMC11894192 DOI: 10.1038/s41418-024-01377-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024] Open
Abstract
Upregulation of mitochondrial respiration coupled with high ROS-scavenging capacity is a characteristic shared by drug-tolerant cells in several cancers. As translational fidelity is essential for cell fitness, protection of the mitochondrial and cytosolic ribosomes from oxidative damage is pivotal. While mechanisms for recognising and repairing such damage exist in the cytoplasm, the corresponding process in the mitochondria remains unclear.By performing Ascorbate PEroXidase (APEX)-proximity ligation assay directed to the mitochondrial matrix followed by isolation and sequencing of RNA associated to mitochondrial proteins, we identified the nuclear-encoded lncRNA ROSALIND as an interacting partner of ribosomes. ROSALIND is upregulated in recurrent tumours and its expression can discriminate between responders and non-responders to immune checkpoint blockade in a melanoma cohort of patients. Featuring an unusually high G content, ROSALIND serves as a substrate for oxidation. Consequently, inhibiting ROSALIND leads to an increase in ROS and protein oxidation, resulting in severe mitochondrial respiration defects. This, in turn, impairs melanoma cell viability and increases immunogenicity both in vitro and ex vivo in preclinical humanised cancer models. These findings underscore the role of ROSALIND as a novel ROS buffering system, safeguarding mitochondrial translation from oxidative stress, and shed light on potential therapeutic strategies for overcoming cancer therapy resistance.
Collapse
Affiliation(s)
- Vicky Katopodi
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Alessandro Marino
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Nikoleta Pateraki
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Yvessa Verheyden
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sonia Cinque
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Elena Lara Jimenez
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sara Adnane
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ewout Demesmaeker
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Alice Scomparin
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Rita Derua
- Laboratory for Protein Phosphorylation and Proteomics, Leuven, Belgium
| | - Elisabetta Groaz
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Leuven, Belgium
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium.
- Trace, Leuven Cancer Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Wei S, Zhi Y, Chen Z, Zhang Y, Duan K, Li M, Lv G. The role of the glutathione pathway in tracheal regeneration with aortic allografts through antioxidant-driven tissue integration. Commun Biol 2025; 8:332. [PMID: 40021729 PMCID: PMC11871065 DOI: 10.1038/s42003-025-07741-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
Tracheal regeneration remains a major challenge due to the lack of efficient graft integration and functional restoration. Current approaches fail to address oxidative stress-induced tissue remodeling. Here, we show that the glutathione pathway plays a pivotal role in tracheal regeneration with aortic allografts by modulating redox homeostasis and promoting host-graft integration. Through transcriptomic profiling, histological analyses, and functional assessment, we demonstrate that antioxidant-driven tissue remodeling enhances epithelialization, neovascularization, and extracellular matrix organization, thereby improving graft stability and biomechanical properties. These findings provide mechanistic insights into oxidative stress-mediated tissue remodeling and suggest that targeting redox signaling could optimize bioengineered tracheal grafts for clinical translation.
Collapse
Affiliation(s)
- Shixiong Wei
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 130021, Changchun, China
- Department of Thoracic Surgery, The First Hospital of Jilin University, 130021, Changchun, China
- Medicine & Engineering & Informatics Fusion and Transformation Key Laboratory of Luzhou City, 646000, Luzhou, China
| | - Yao Zhi
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 130021, Changchun, China
| | - Zhou Chen
- Institute of Atomic and Molecular Physics, Jilin University, 130012, Changchun, China
- Advanced Light Field and Modern Medical Treatment Science and Technology Innovation Center of Jilin Province, Jilin University, 130012, Changchun, China
| | - Yiyuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 130021, Changchun, China
- Department of Thoracic Surgery, The First Hospital of Jilin University, 130021, Changchun, China
| | - Kexing Duan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 130021, Changchun, China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 130021, Changchun, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 130021, Changchun, China.
| |
Collapse
|
13
|
Long Y, Shi H, Ye J, Qi X. Exploring Strategies to Prevent and Treat Ovarian Cancer in Terms of Oxidative Stress and Antioxidants. Antioxidants (Basel) 2025; 14:114. [PMID: 39857448 PMCID: PMC11762571 DOI: 10.3390/antiox14010114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Oxidative stress is a state of imbalance between the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) and the antioxidant defence system in the body. Oxidative stress may be associated with a variety of diseases, such as ovarian cancer, diabetes mellitus, and neurodegeneration. The generation of oxidative stress in ovarian cancer, one of the common and refractory malignancies among gynaecological tumours, may be associated with several factors. On the one hand, the increased metabolism of ovarian cancer cells can lead to the increased production of ROS, and on the other hand, the impaired antioxidant defence system of ovarian cancer cells is not able to effectively scavenge the excessive ROS. In addition, chemotherapy and radiotherapy may elevate the oxidative stress in ovarian cancer cells. Oxidative stress can cause oxidative damage, promote the development of ovarian cancer, and even result in drug resistance. Therefore, studying oxidative stress in ovarian cancer is important for the prevention and treatment of ovarian cancer. Antioxidants, important markers of oxidative stress, might serve as one of the strategies for preventing and treating ovarian cancer. In this review, we will discuss the complex relationship between oxidative stress and ovarian cancer, as well as the role and therapeutic potential of antioxidants in ovarian cancer, thus guiding future research and clinical interventions.
Collapse
Affiliation(s)
| | | | | | - Xiaorong Qi
- Key Laboratory of Birth, Defects and Related Diseases of Women and Children, Department of Gynecology and Obstetrics, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, China; (Y.L.); (H.S.); (J.Y.)
| |
Collapse
|
14
|
Yang C, Fu J, Zheng F, Fu Y, Duan X, Zuo R, Zhu J. Aconitine promotes ROS-activated P38/MAPK/Nrf2 pathway to inhibit autophagy and promote myocardial injury. J Cardiothorac Surg 2024; 19:665. [PMID: 39707526 DOI: 10.1186/s13019-024-03149-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Aconitine has cardiotoxicity, but the mechanism of cardiotoxicity induced by aconitine is limited. The aim of this study was to investigate the mechanism of myocardial injury induced by aconitine. METHODS Using aconitine, ROS inhibitor N-acetylcysteine(NAC), the autophagy activitor Rapamycin (Rap) or the P38/MAPK pathway activitor Dehydrocorydaline treats H9C2 cells. CCK-8 assay was used to assay cell proliferation activity. Flow Cytometry was used to detect cell apoptosis. Dichloro-dihydrofluorescein diacetate was used to detect ROS levels. The expression of LC3 was detected by Immunofluorescence Staining. Western blotting detected the expression of related proteins. The mRNA levels of inflammatory factors were detected by RT-qPCR. RESULTS Aconitine inhibits cardiomyocyte proliferation, induces apoptosis and secretion of inflammatory factors. Aconitine activates the P38/MAPK/Nrf2 pathway, induces ROS increase, and promotes autophagy. NAC can inhibit proliferation inhibition, apoptosis, inflammation and P38/MAPK/Nrf2 pathway activation induced by aconitine. Rap and P38 activators can partially recover the effects of NAC on proliferation, apoptosis, inflammation and autophagy of cardiomyocytes. CONCLUSION Aconitine promotes ROS-activated P38/MAPK/Nrf2 pathway to inhibit autophagy and promote myocardial injury.
Collapse
Affiliation(s)
- Chunai Yang
- Department of Emergency, The Affiliated Hospital of Yunnan University, Kunming, 650021, China
| | - Jinxiao Fu
- Department of Geriatric Medicine, The Affiliated Hospital of Yunnan University, 176 Qingnian Road, Wuhua District, Kunming, 650021, Yunnan, China.
| | - Fenshuang Zheng
- Department of Emergency, The Affiliated Hospital of Yunnan University, Kunming, 650021, China
| | - Yangshan Fu
- Department of Emergency, The Affiliated Hospital of Yunnan University, Kunming, 650021, China
| | - Xueqiong Duan
- Department of Emergency, The Affiliated Hospital of Yunnan University, Kunming, 650021, China
| | - Ruiling Zuo
- Department of Emergency, The Affiliated Hospital of Yunnan University, Kunming, 650021, China
| | - Junbo Zhu
- Department of Emergency, The Affiliated Hospital of Yunnan University, Kunming, 650021, China
| |
Collapse
|
15
|
Woldeselassie M, Tamene A. Therapeutic controversies over use of antioxidant supplements during cancer treatment: a scoping review. Front Nutr 2024; 11:1480780. [PMID: 39717397 PMCID: PMC11663640 DOI: 10.3389/fnut.2024.1480780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/21/2024] [Indexed: 12/25/2024] Open
Abstract
Background Antioxidant supplements are widely used during cancer treatment to prevent oxidative stress, reduce treatment toxicities, and improve patient outcomes. However, current literature reveals significant gaps suggesting that antioxidants may protect both healthy and tumor cells from oxidative damage, thereby reducing treatment efficacy. It is for this reason that antioxidant supplements have become a source of therapeutic controversy. Objective To review therapeutic controversies over the use of antioxidant supplements during cancer treatment. Methods Scoping review of the international published articles following the Arksey and O'Malley framework, cross-sectional studies, clinical and pre-clinical studies, systematic and umbrella reviews and grey literatures published from 2014 to 2024 with all age patient populations were included. A structured literature search was conducted of CINAHL, EMBASE, MEDLINE, Google Scholar, using key medical subject heading words and Cochrane Collaboration and Joanna Briggs Institute databases. All included studies were reviewed independently by two investigators. Data were extracted, collated by type of antioxidants, summarized in tables and synthesized for analysis. Result A total of 1, 550 articles were identified. After reviewing all literatures, twenty-one (21) were full-text articles, grey literatures (2), and systematic reviews (42) and umbrella reviews (3), met the criteria for inclusion. In this review, the use of antioxidant supplements can benefit cancer cells in the same way as they do for normal cells during cancer treatment. In addition, not all antioxidants were effective in inhibiting oxidative stress, reduce treatment toxicities, and improve patient outcomes. Conclusion and recommendations According to this review, the use of antioxidant supplements can benefit tumor cells in the same manner as they do for normal cells. Therefore, oncologists should advise not to take antioxidant supplements during chemotherapy and/or radiotherapy. Future research including potential clinical and preclinical trials, mechanistic studies, and exploration of different vitamin and mineral supplement studies are required to uncover the complete potential of antioxidant supplements for cancer treatment or determine their safety and effectiveness when used alongside standard cancer treatments. Furthermore, the results of this review could be used for future systematic review of therapeutic controversies over use of antioxidant supplements during cancer treatment.
Collapse
Affiliation(s)
| | - Aynadis Tamene
- College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
- Food Science and Nutrition, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
16
|
O'Reilly A, Zhao W, Wickström S, Arnér ESJ, Kiessling R. Reactive oxygen species: Janus-faced molecules in the era of modern cancer therapy. J Immunother Cancer 2024; 12:e009409. [PMID: 39645234 PMCID: PMC11629020 DOI: 10.1136/jitc-2024-009409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024] Open
Abstract
Oxidative stress, that is, an unbalanced increase in reactive oxygen species (ROS), contributes to tumor-induced immune suppression and limits the efficacy of immunotherapy. Cancer cells have inherently increased ROS production, intracellularly through metabolic perturbations and extracellularly through activation of NADPH oxidases, which promotes cancer progression. Further increased ROS production or impaired antioxidant systems, induced, for example, by chemotherapy or radiotherapy, can preferentially kill cancer cells over healthy cells. Inflammatory cell-derived ROS mediate immunosuppressive effects of myeloid-derived suppressor cells and activated granulocytes, hampering antitumor effector cells such as T cells and natural killer (NK) cells. Cancer therapies modulating ROS levels in tumors may thus have entirely different consequences when targeting cancer cells versus immune cells. Here we discuss the possibility of developing more efficient cancer therapies based on reduction-oxidation modulation, as either monotherapies or in combination with immunotherapy. Short-term, systemic administration of antioxidants or drugs blocking ROS production can boost the immune system and act in synergy with immunotherapy. However, prolonged use of antioxidants can instead enhance tumor progression. Alternatives to systemic antioxidant administration are under development where gene-modified or activated T cells and NK cells are shielded ex vivo against the harmful effects of ROS before the infusion to patients with cancer.
Collapse
Affiliation(s)
- Aine O'Reilly
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, University College Cork, Cork, Ireland
- The Christie NHS Foundation Trust, Manchester, UK
| | - Wenchao Zhao
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Stina Wickström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
17
|
Hao X, Chen X, Ren C, Pan Y, Xu Z, Wang Q, Liu X. Association between composite dietary antioxidant index and erectile dysfunction: a cross-sectional study from NHANES. BMC Public Health 2024; 24:3362. [PMID: 39623347 PMCID: PMC11613466 DOI: 10.1186/s12889-024-20880-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Individual antioxidants may not fully capture the comprehensive antioxidant intake from an individual's diet. This study utilizes the Complex Dietary Antioxidant Index (CDAI) to evaluate the combined effects of various dietary antioxidants in the diet. The goal is to investigate the relationship between CDAI and the incidence of erectile dysfunction (ED), offering insights for dietary guidelines and intervention strategies aimed at mitigating the burden of ED. METHODS This cross-sectional study utilized data from the National Health and Nutrition Examination Survey (NHANES) database in the years 2001-2004. We employed a weighted multivariate logistic regression model to validate the relationship between CDAI and ED. Subgroup analyses were conducted to explore the correlation between CDAI and ED across different subgroups. Additionally, we used propensity score matching (PSM) to adjust for several key confounding variables, enhancing the robustness of the results. RESULTS In the fully adjusted multivariate logistic regression model for confounding variables, CDAI is negatively correlated with the risk of ED (OR = 0.95, 95% CI: 0.92-0.99, P = 0.005). When CDAI is transformed into a categorical variable based on quartiles, Q3 (OR = 0.73, 95% CI: 0.53-0.99, P = 0.040) and Q4 (OR = 0.70, 95% CI: 0.51-0.96, P = 0.026) show a negative correlation with the risk of ED. Subgroup analysis reveals no significant interaction. After adjusting for major confounding variables through PSM, the association between CDAI and reduced risk of ED remains significant. CONCLUSION In our study cohort, there is an association between CDAI and a reduced risk of ED, and further research is needed to validate and refine this conclusion.
Collapse
Affiliation(s)
- Xuexue Hao
- Department of Urology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xiangyu Chen
- Department of Urology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Congzhe Ren
- Department of Urology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yang Pan
- Department of Urology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Zhunan Xu
- Department of Urology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Qihua Wang
- Department of Urology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
18
|
Chen Z, Yu L, Zheng Z, Wang X, Guo Q, Chen Y, Zhang Y, Zhang Y, Xiao J, Chen K, Fan H, Ding Y. CPT1A mediates radiation sensitivity in colorectal cancer. eLife 2024; 13:RP97827. [PMID: 39607749 PMCID: PMC11604221 DOI: 10.7554/elife.97827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
The prevalence and mortality rates of colorectal cancer (CRC) are increasing worldwide. Radiation resistance hinders radiotherapy, a standard treatment for advanced CRC, leading to local recurrence and metastasis. Elucidating the molecular mechanisms underlying radioresistance in CRC is critical to enhance therapeutic efficacy and patient outcomes. Bioinformatic analysis and tumour tissue examination were conducted to investigate the CPT1A mRNA and protein levels in CRC and their correlation with radiotherapy efficacy. Furthermore, lentiviral overexpression and CRISPR/Cas9 lentiviral vectors, along with in vitro and in vivo radiation experiments, were used to explore the effect of CPT1A on radiosensitivity. Additionally, transcriptomic sequencing, molecular biology experiments, and bioinformatic analyses were employed to elucidate the molecular mechanisms by which CPT1A regulates radiosensitivity. CPT1A was significantly downregulated in CRC and negatively correlated with responsiveness to neoadjuvant radiotherapy. Functional studies suggested that CPT1A mediates radiosensitivity, influencing reactive oxygen species (ROS) scavenging and DNA damage response. Transcriptomic and molecular analyses highlighted the involvement of the peroxisomal pathway. Mechanistic exploration revealed that CPT1A downregulates the FOXM1-SOD1/SOD2/CAT axis, moderating cellular ROS levels after irradiation and enhancing radiosensitivity. CPT1A downregulation contributes to radioresistance in CRC by augmenting the FOXM1-mediated antioxidant response. Thus, CPT1A is a potential biomarker of radiosensitivity and a novel target for overcoming radioresistance, offering a future direction to enhance CRC radiotherapy.
Collapse
Affiliation(s)
- Zhenhui Chen
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhouChina
| | - Lu Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Zhihao Zheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Xusheng Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Qiqing Guo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yuchuan Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Department of Infectious Diseases, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yaowei Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yuqin Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Jianbiao Xiao
- Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical UniversityGuangzhouChina
| | - Keli Chen
- HuiQiao Medical Center, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Hongying Fan
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhouChina
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
19
|
Lin YC, Ku CC, Wuputra K, Wu DC, Yokoyama KK. Vulnerability of Antioxidant Drug Therapies on Targeting the Nrf2-Trp53-Jdp2 Axis in Controlling Tumorigenesis. Cells 2024; 13:1648. [PMID: 39404411 PMCID: PMC11475825 DOI: 10.3390/cells13191648] [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: 08/15/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Control of oxidation/antioxidation homeostasis is important for cellular protective functions, and disruption of the antioxidation balance by exogenous and endogenous ligands can lead to profound pathological consequences of cancerous commitment within cells. Although cancers are sensitive to antioxidation drugs, these drugs are sometimes associated with problems including tumor resistance or dose-limiting toxicity in host animals and patients. These problems are often caused by the imbalance between the levels of oxidative stress-induced reactive oxygen species (ROS) and the redox efficacy of antioxidants. Increased ROS levels, because of abnormal function, including metabolic abnormality and signaling aberrations, can promote tumorigenesis and the progression of malignancy, which are generated by genome mutations and activation of proto-oncogene signaling. This hypothesis is supported by various experiments showing that the balance of oxidative stress and redox control is important for cancer therapy. Although many antioxidant drugs exhibit therapeutic potential, there is a heterogeneity of antioxidation functions, including cell growth, cell survival, invasion abilities, and tumor formation, as well as the expression of marker genes including tumor suppressor proteins, cell cycle regulators, nuclear factor erythroid 2-related factor 2, and Jun dimerization protein 2; their effectiveness in cancer remains unproven. Here, we summarize the rationale for the use of antioxidative drugs in preclinical and clinical antioxidant therapy of cancer, and recent advances in this area using cancer cells and their organoids, including the targeting of ROS homeostasis.
Collapse
Affiliation(s)
- Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kazunari K. Yokoyama
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
20
|
Qian Z, Sun C, Li Q, Xie Y, Zhan L, Liu X, Wang G, Wei Y, Qiu J, Peng Q. Unravelling the antioxidant behaviour of self-assembly β-Sheet in silk fibroin. Redox Biol 2024; 76:103307. [PMID: 39213701 PMCID: PMC11401358 DOI: 10.1016/j.redox.2024.103307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Local oxidative stress in diseases or injury severely hinders cell homeostasis and organ regeneration. Antioxidant therapy is an effective strategy for oxidative stress treatment. Biomaterials with good biocompatibility and reactive oxygen species (ROS) scavenging ability are good choices for antioxidant therapeutics. However, there are few natural biomaterials that are identified with both biocompatibility and strong antioxidant activity. Here, we show, for the first time, that silk fibroin (SF) is a strong antioxidant, which can eliminate ROS in both cells and zebrafish. We further demonstrate that the β-sheet structures turn into a random coiled structure when SF is treated with hydrogen peroxide. The content of β-sheet structures can be increased by heating, thus enhancing the antioxidation properties of SF. Therefore, SF can serve as a good antioxidant biomaterial for therapeutics, and its β-sheet structure-based antioxidation mechanism provides a novel theoretical basis, which could be a new cue for more antioxidant biomaterial discovery and identification.
Collapse
Affiliation(s)
- Zhiyong Qian
- Department of Anatomy the Basic Medicine College, Inner Mongolia Medical University, Hohhot, 010000, Inner Mongolia, China; Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Chang Sun
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Qianqian Li
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Yafan Xie
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Lingpeng Zhan
- Institute for Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Xiangli Liu
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Guanbo Wang
- Institute for Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China; School of Materials Science and Engineering, North Minzu University, Yinchuan, 750021, China.
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| | - Qin Peng
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
21
|
Wang M, Xu Z, Wang Z, Xu X, Sun Y. Analysis of the mechanism of berberine against stomach carcinoma based on network pharmacology and experimental validation. Transl Cancer Res 2024; 13:4593-4607. [PMID: 39430861 PMCID: PMC11483340 DOI: 10.21037/tcr-24-668] [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: 04/23/2024] [Accepted: 08/14/2024] [Indexed: 10/22/2024]
Abstract
Background Although the therapeutic effects of berberine have received some attention in recent years, its potential mechanisms underlying its action against stomach carcinoma (SC) remain unclear. In this study, we aimed to elucidate the mechanisms underlying the effects of berberine against SC using a network pharmacology and experimental verification approach. Methods Several publicly available databases were used to collect the targets of berberine and SC. Protein-protein interaction (PPI) network, enrichment analyses and molecular docking were performed based on the potential targets of berberine against SC. The potential clinical significance and prognostic value of the targets were predicted by using nomogram and receiver operating characteristic (ROC) analyses. Then the viability and apoptosis of SC cells treated with berberine were determined. Moreover, reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and adenosine triphosphate (ATP) measurements and western blot assay were carried out to validate the predicted mechanisms. Results Seventy-six potential targets of berberine against SC were identified. The construction of PPI network enabled the identification of hub targets, such as AKT1, TP53, IL6, JUN and MAPK1. Enrichment analyses showed that berberine was involved in apoptosis, mitophagy, ROS metabolic process, AMPK and MAPK signaling pathway. The expression levels of hub targets also contributed to the clinical prognosis of patients with SC. Molecular docking revealed the possible patterns of direct interaction between berberine and target proteins, including AMPK, TP53 and MAPK1. Experimental results showed that berberine reduced SC cell viability, promoted apoptosis and ROS generation, and contributed to reductions in MMP and ATP levels. Western blot assay demonstrated that berberine increased AMPK and TP53 expression, while decreased phosphorylated-MAPK3/1 expression. Conclusions We elucidated the potential action mechanisms of berberine against SC using a network pharmacology approach. Some predicted mechanisms underlying the anti-SC effects were verified based on experimental approaches. Our findings provide a meaningful foundation for berberine as a cellular apoptosis-inducing and energy metabolism-regulating agent against SC. However, in vivo experiments and clinical studies need to be further carried out. Moreover, it is necessary to study the potential negative effects of berberine thoroughly.
Collapse
Affiliation(s)
- Meng Wang
- Department of Traditional Chinese Medicine Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
- Traditional Chinese Medicine Oncology Teaching and Research Office, Department of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| | - Zeyu Xu
- Department of Cardiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziyang Wang
- Department of Cardiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaowan Xu
- Department of Traditional Chinese Medicine Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
- Traditional Chinese Medicine Oncology Teaching and Research Office, Department of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| | - Yongning Sun
- Department of Cardiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
22
|
Xiang H, Shen B, Zhang C, Li R. Bioactive Nanoliposomes for Enhanced Sonodynamic-Triggered Disulfidptosis-Like Cancer Cell Death via Lipid Peroxidation. Int J Nanomedicine 2024; 19:8929-8947. [PMID: 39246429 PMCID: PMC11379027 DOI: 10.2147/ijn.s464178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction Cell death regulation holds a unique value in the field of cancer therapy. Recently, disulfidptosis has garnered substantial scientific attention. Previous studies have reported that sonodynamic therapy (SDT) based on reactive oxygen species (ROS) can regulate cancer cell death, achieving an limited anti-cancer effect. However, the integration of SDT with disulfidptosis as an anti-cancer strategy has not been extensively developed. In this study, we constructed an artificial membrane disulfidptosis sonosensitizer, specifically, a nanoliposome (SC@lip) coated with a combination of the chemotherapy medicine Sorafenib (Sora) and sonosensitizer Chlorin e6 (Ce6), to realize a one-stop enhanced SDT effect that induces disulfidptosis-like cancer cell death. Methods Sorafenib and Ce6 were co-encapsulated into PEG-modified liposomes, and SC@Lip was constructed using a simple rotary evaporation phacoemulsification method. The cell phagocytosis, ROS generation ability, glutathione (GSH) depletion ability, lipid peroxidation (LPO), and disulfidptosis-like death mediated by SC@Lip under ultrasound (US) irradiation were evaluated. Based on a 4T1 subcutaneous tumor model, both the in vivo biological safety assessment and the efficacy of SDT were assessed. Results SC@Lip exhibits high efficiency in cellular phagocytosis. After being endocytosed by 4T1 cells, abundant ROS were produced under SDT activation, and the cell survival rates were below 5%. When applied to a 4T1 subcutaneous tumor model, the enhanced SDT mediated by SC@Lip inhibited tumor growth and prolonged the survival time of mice. In vitro and in vivo experiments show that SC@Lip can enhance the SDT effect and trigger disulfidptosis-like cancer cell death, thus achieving anti-tumor efficacy both in vitro and in vivo. Conclusion SC@Lip is a multifunctional nanoplatform with an artificial membrane, which can integrate the functions of sonosensitization and GSH depletion into a biocompatible nanoplatform, and can be used to enhance the SDT effect and promote disulfidptosis-like cancer cell death.
Collapse
Affiliation(s)
- Hongwei Xiang
- Department of Ultrasound, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Bin Shen
- Department of Ultrasound, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Chunmei Zhang
- Department of Ultrasound, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Rui Li
- Department of Ultrasound, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
23
|
Zheng Y, Ma Y, Xiong Q, Zhu K, Weng N, Zhu Q. The role of artificial intelligence in the development of anticancer therapeutics from natural polyphenols: Current advances and future prospects. Pharmacol Res 2024; 208:107381. [PMID: 39218422 DOI: 10.1016/j.phrs.2024.107381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/06/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Natural polyphenols, abundant in the human diet, are derived from a wide variety of sources. Numerous preclinical studies have demonstrated their significant anticancer properties against various malignancies, making them valuable resources for drug development. However, traditional experimental methods for developing anticancer therapies from natural polyphenols are time-consuming and labor-intensive. Recently, artificial intelligence has shown promising advancements in drug discovery. Integrating AI technologies into the development process for natural polyphenols can substantially reduce development time and enhance efficiency. In this study, we review the crucial roles of natural polyphenols in anticancer treatment and explore the potential of AI technologies to aid in drug development. Specifically, we discuss the application of AI in key stages such as drug structure prediction, virtual drug screening, prediction of biological activity, and drug-target protein interaction, highlighting the potential to revolutionize the development of natural polyphenol-based anticancer therapies.
Collapse
Affiliation(s)
- Ying Zheng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Yifei Ma
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Qunli Xiong
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Kai Zhu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350011, PR China
| | - Ningna Weng
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350011, PR China
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China.
| |
Collapse
|
24
|
Feng Y, Zhang Y, Li H, Wang T, Lu F, Liu R, Xie G, Song L, Huang B, Li X, Ding Y, Yang J, Jia Z, Huang Z. Enzalutamide inhibits PEX10 function and sensitizes prostate cancer cells to ROS activators. Cell Death Dis 2024; 15:559. [PMID: 39097593 PMCID: PMC11297951 DOI: 10.1038/s41419-024-06937-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/05/2024]
Abstract
Sharply increased reactive oxygen species (ROS) are thought to induce oxidative stress, damage cell structure and cause cell death; however, its role in prostate cancer remains unclear. Enzalutamide is a widely used anti-prostate cancer drug that antagonizes androgen binding with its receptor. Further exploration of the mechanism and potential application strategies of enzalutamide is crucial for the treatment of prostate cancer. Here, we confirmed PEX10 can be induced by ROS activators while reduce ROS level in prostate cancer cells, which weakened the anti-tumor effect of ROS activators. The androgen receptor (AR) can promote the expression of PEX10 by acting as an enhancer in cooperation with FOXA1. The anti-tumor drug enzalutamide inhibits PEX10 by inhibiting the function of AR, and synergize with ROS activators ML210 or RSL3 to produce a stronger anti-tumor effect, thereby sensitizing cells to ROS activators. This study reveals a previously unrecognized function of enzalutamide and AR by regulating PEX10 and suggests a new strategy of enzalutamide application in prostate cancer treatment.
Collapse
Affiliation(s)
- Yuankang Feng
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yu Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Tao Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fubo Lu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ruoyang Liu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Guoqing Xie
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Liang Song
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Budeng Huang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiang Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yinghui Ding
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jinjian Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhankui Jia
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Zhenlin Huang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
25
|
Kalinina E. Glutathione-Dependent Pathways in Cancer Cells. Int J Mol Sci 2024; 25:8423. [PMID: 39125992 PMCID: PMC11312684 DOI: 10.3390/ijms25158423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
The most abundant tripeptide-glutathione (GSH)-and the major GSH-related enzymes-glutathione peroxidases (GPxs) and glutathione S-transferases (GSTs)-are highly significant in the regulation of tumor cell viability, initiation of tumor development, its progression, and drug resistance. The high level of GSH synthesis in different cancer types depends not only on the increasing expression of the key enzymes of the γ-glutamyl cycle but also on the changes in transport velocity of its precursor amino acids. The ability of GPxs to reduce hydroperoxides is used for cellular viability, and each member of the GPx family has a different mechanism of action and site for maintaining redox balance. GSTs not only catalyze the conjugation of GSH to electrophilic substances and the reduction of organic hydroperoxides but also take part in the regulation of cellular signaling pathways. By catalyzing the S-glutathionylation of key target proteins, GSTs are involved in the regulation of major cellular processes, including metabolism (e.g., glycolysis and the PPP), signal transduction, transcription regulation, and the development of resistance to anticancer drugs. In this review, recent findings in GSH synthesis, the roles and functions of GPxs, and GST isoforms in cancer development are discussed, along with the search for GST and GPx inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Elena Kalinina
- T.T. Berezov Department of Biochemistry, Peoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
| |
Collapse
|
26
|
Wang S, Mao Y, Rong S, Liu G, Cao Y, Yang Z, Yu H, Zhang X, Fang H, Cai Z, Chen Y, Huang H, Li H. Engineering Magnetic Extracellular Vesicles Mimetics for Enhanced Targeting Chemodynamic Therapy to Overcome Ovary Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:39021-39034. [PMID: 39033517 DOI: 10.1021/acsami.4c06862] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Chemodynamic therapy (CDT), employing metal ions to transform endogenous H2O2 into lethal hydroxyl radicals (•OH), has emerged as an effective approach for tumor treatment. Yet, its efficacy is diminished by glutathione (GSH), commonly overexpressed in tumors. Herein, a breakthrough strategy involving extracellular vesicle (EV) mimetic nanovesicles (NVs) encapsulating iron oxide nanoparticles (IONPs) and β-Lapachone (Lapa) was developed to amplify intracellular oxidative stress. The combination, NV-IONP-Lapa, created through a serial extrusion from ovarian epithelial cells showed excellent biocompatibility and leveraged magnetic guidance to enhance endocytosis in ovarian cancer cells, resulting in selective H2O2 generation through Lapa catalysis by NADPH quinone oxidoreductase 1 (NQO1). Meanwhile, the iron released from IONPs ionization under acidic conditions triggered the conversion of H2O2 into •OH by the Fenton reaction. Additionally, the catalysis process of Lapa eliminated GSH in tumor, further amplifying oxidative stress. The designed NV-IONP-Lapa demonstrated exceptional tumor targeting, facilitating MR imaging, and enhanced tumor suppression without significant side effects. This study presents a promising NV-based drug delivery system for exploiting CDT against NQO1-overexpressing tumors by augmenting intratumoral oxidative stress.
Collapse
Affiliation(s)
- Shuai Wang
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yinghua Mao
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
| | - Shu Rong
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
| | - Guangquan Liu
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210001, China
| | - Yongping Cao
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
| | - Zhan Yang
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
| | - Huanhuan Yu
- Department of Clinical Pharmacy, General Hospital of Eastern Theater Command, Nanjing 210002, China
| | - Xinrui Zhang
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
| | - Hongyue Fang
- Department of Third Outpatient, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhipeng Cai
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
| | - Yonghong Chen
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
| | - Hao Huang
- Department of Obstetrics and Gynecology, Foshan Fosun Chancheng Hospital, Foshan 528000, China
| | - Hong Li
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
| |
Collapse
|
27
|
Li H, Gong Q, Luo K. Biomarker-driven molecular imaging probes in radiotherapy. Theranostics 2024; 14:4127-4146. [PMID: 38994026 PMCID: PMC11234278 DOI: 10.7150/thno.97768] [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: 04/25/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
Background: Biomarker-driven molecular imaging has emerged as an integral part of cancer precision radiotherapy. The use of molecular imaging probes, including nanoprobes, have been explored in radiotherapy imaging to precisely and noninvasively monitor spatiotemporal distribution of biomarkers, potentially revealing tumor-killing mechanisms and therapy-induced adverse effects during radiation treatment. Methods: We summarized literature reports from preclinical studies and clinical trials, which cover two main parts: 1) Clinically-investigated and emerging imaging biomarkers associated with radiotherapy, and 2) instrumental roles, functions, and activatable mechanisms of molecular imaging probes in the radiotherapy workflow. In addition, reflection and future perspectives are proposed. Results: Numerous imaging biomarkers have been continuously explored in decades, while few of them have been successfully validated for their correlation with radiotherapeutic outcomes and/or radiation-induced toxicities. Meanwhile, activatable molecular imaging probes towards the emerging biomarkers have exhibited to be promising in animal or small-scale human studies for precision radiotherapy. Conclusion: Biomarker-driven molecular imaging probes are essential for precision radiotherapy. Despite very inspiring preliminary results, validation of imaging biomarkers and rational design strategies of probes await robust and extensive investigations. Especially, the correlation between imaging biomarkers and radiotherapeutic outcomes/toxicities should be established through multi-center collaboration involving a large cohort of patients.
Collapse
Affiliation(s)
- Haonan Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, 699 Jinyuan Xi Road, Jimei District, 361021 Xiamen, Fujian, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
28
|
Zhou H, He J, Liu R, Cheng J, Yuan Y, Mao W, Zhou J, He H, Liu Q, Tan W, Shuai C, Deng Y. Microenvironment-responsive metal-phenolic network release platform with ROS scavenging, anti-pyroptosis, and ECM regeneration for intervertebral disc degeneration. Bioact Mater 2024; 37:51-71. [PMID: 38515609 PMCID: PMC10954684 DOI: 10.1016/j.bioactmat.2024.02.036] [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: 12/14/2023] [Revised: 02/13/2024] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) can be caused by aging, injury, and genetic factors. The pathological changes associated with IVDD include the excessive accumulation of reactive oxygen species (ROS), cellular pyroptosis, and extracellular matrix (ECM) degradation. There are currently no approved specific molecular therapies for IVDD. In this study, we developed a multifunctional and microenvironment-responsive metal-phenolic network release platform, termed TMP@Alg-PBA/PVA, which could treat (IL-1β)-induced IVDD. The metal-phenolic network (TA-Mn-PVP, TMP) released from this platform targeted mitochondria to efficiently scavenge ROS and reduce ECM degradation. Pyroptosis was suppressed through the inhibition of the IL-17/ERK signaling pathway. These findings demonstrate the versatility of the platform. And in a rat model of IVDD, TMP@Alg-PBA/PVA exhibited excellent therapeutic effects by reducing the progression of the disease. TMP@Alg-PBA/PVA, therefore, presents clinical potential for the treatment of IVDD.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
- Department of Joint Surgery and Sports Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Jinpeng He
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Renfeng Liu
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Jun Cheng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Yuhao Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Wanpu Mao
- Department of Joint Surgery and Sports Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Jun Zhou
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Honghui He
- Department of Joint Surgery and Sports Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Qianqi Liu
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Wei Tan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Cijun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| |
Collapse
|
29
|
Gu X, Mu C, Zheng R, Zhang Z, Zhang Q, Liang T. The Cancer Antioxidant Regulation System in Therapeutic Resistance. Antioxidants (Basel) 2024; 13:778. [PMID: 39061847 PMCID: PMC11274344 DOI: 10.3390/antiox13070778] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/15/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Antioxidants play a pivotal role in neutralizing reactive oxygen species (ROS), which are known to induce oxidative stress. In the context of cancer development, cancer cells adeptly maintain elevated levels of both ROS and antioxidants through a process termed "redox reprogramming". This balance optimizes the proliferative influence of ROS while simultaneously reducing the potential for ROS to cause damage to the cell. In some cases, the adapted antioxidant machinery can hamper the efficacy of treatments for neoplastic diseases, representing a significant facet of the resistance mechanisms observed in cancer therapy. In this review, we outline the contribution of antioxidant systems to therapeutic resistance. We detail the fundamental constituents of these systems, encompassing the central regulatory mechanisms involving transcription factors (of particular importance is the KEAP1/NRF2 signaling axis), the molecular effectors of antioxidants, and the auxiliary systems responsible for NADPH generation. Furthermore, we present recent clinical trials based on targeted antioxidant systems for the treatment of cancer, assessing the potential as well as challenges of this strategy in cancer therapy. Additionally, we summarize the pressing issues in the field, with the aim of illuminating a path toward the emergence of novel anticancer therapeutic approaches by orchestrating redox signaling.
Collapse
Affiliation(s)
- Xuanhao Gu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Chunyang Mu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Rujia Zheng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Zhe Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
30
|
Chu T, Liu Y, Gao Y, Zhou C, Huang W, Zheng Y. Colorimetric array sensor based on bimetallic nitrogen-doped carbon-based nanozyme material to detect multiple antioxidants. Mikrochim Acta 2024; 191:365. [PMID: 38831060 DOI: 10.1007/s00604-024-06444-8] [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: 03/19/2024] [Accepted: 05/18/2024] [Indexed: 06/05/2024]
Abstract
Copper-cobalt bimetallic nitrogen-doped carbon-based nanoenzymatic materials (CuCo@NC) were synthesized using a one-step pyrolysis process. A three-channel colorimetric sensor array was constructed for the detection of seven antioxidants, including cysteine (Cys), uric acid (UA), tea polyphenols (TP), lysine (Lys), ascorbic acid (AA), glutathione (GSH), and dopamine (DA). CuCo@NC with peroxidase activity was used to catalyze the oxidation of TMB by H2O2 at three different ratios of metal sites. The ability of various antioxidants to reduce the oxidation products of TMB (ox TMB) varied, leading to distinct absorbance changes. Linear discriminant analysis (LDA) results showed that the sensor array was capable of detecting seven antioxidants in buffer and serum samples. It could successfully discriminate antioxidants with a minimum concentration of 10 nM. Thus, multifunctional sensor arrays based on CuCo@NC bimetallic nanoenzymes not only offer a promising strategy for identifying various antioxidants but also expand their applications in medical diagnostics and environmental analysis of food.
Collapse
Affiliation(s)
- Tingting Chu
- Hubei Key Laboratory of Selenium Resources Research and Biological Applications, Hubei Minzu University, Enshi City, Hubei, China
- Institute of Selenium Science and Industry, Hubei Minzu University, Enshi City, Hubei, China
- School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi, 445000, China
| | - Yaopeng Liu
- Hubei Key Laboratory of Selenium Resources Research and Biological Applications, Hubei Minzu University, Enshi City, Hubei, China
- Institute of Selenium Science and Industry, Hubei Minzu University, Enshi City, Hubei, China
- School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi, 445000, China
| | - Yi Gao
- Hubei Key Laboratory of Selenium Resources Research and Biological Applications, Hubei Minzu University, Enshi City, Hubei, China
- Institute of Selenium Science and Industry, Hubei Minzu University, Enshi City, Hubei, China
- School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi, 445000, China
| | - Chengyu Zhou
- Hubei Key Laboratory of Selenium Resources Research and Biological Applications, Hubei Minzu University, Enshi City, Hubei, China
- Institute of Selenium Science and Industry, Hubei Minzu University, Enshi City, Hubei, China
- School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi, 445000, China
| | - Wensheng Huang
- School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi, 445000, China
| | - Yin Zheng
- Hubei Key Laboratory of Selenium Resources Research and Biological Applications, Hubei Minzu University, Enshi City, Hubei, China.
- Institute of Selenium Science and Industry, Hubei Minzu University, Enshi City, Hubei, China.
- School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi, 445000, China.
| |
Collapse
|
31
|
He SY, Liu W, Huang CM, Huang HM, Cao QL, Li YX, Xu YN, Kim NH, Li YH. Enhancing antioxidant levels and mitochondrial function in porcine oocyte maturation and embryonic development through notoginsenoside R1 supplementation. Reprod Domest Anim 2024; 59:e14631. [PMID: 38828566 DOI: 10.1111/rda.14631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
This study examines the impact of Notoginsenoside R1 (NGR1), a compound from Panax notoginseng, on the maturation of porcine oocytes and their embryonic development, focusing on its effects on antioxidant levels and mitochondrial function. This study demonstrates that supplementing in vitro maturation (IVM) medium with NGR1 significantly enhances several biochemical parameters. These include elevated levels of glutathione (GSH), nuclear factor erythrocyte 2-related factor 2 (NRF2) and mRNA expression of catalase (CAT) and GPX. Concurrently, we observed a decrease in reactive oxygen species (ROS) levels and an increase in JC-1 immunofluorescence, mitochondrial distribution, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α) and nuclear NRF2 mRNA levels. Additionally, there was an increase in ATP production and lipid droplets (LDs) immunofluorescence. These biochemical improvements correlate with enhanced embryonic outcomes, including a higher blastocyst rate, increased total cell count, enhanced proliferative capacity and elevated octamer-binding transcription factor 4 (Oct4) and superoxide dismutase 2 (Sod2) gene expression. Furthermore, NGR1 supplementation resulted in decreased apoptosis, reduced caspase 3 (Cas3) and BCL2-Associated X (Bax) mRNA levels and decreased glucose-regulated protein 78 kD (GRP78) immunofluorescence in porcine oocytes undergoing in vitro maturation. These findings suggest that NGR1 plays a crucial role in promoting porcine oocyte maturation and subsequent embryonic development by providing antioxidant levels and mitochondrial protection.
Collapse
Affiliation(s)
- Sheng-Yan He
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Wen Liu
- Laboratory Animal Center, Southern Medical University, Guangzhou, China
- Department of Animal Science, Chungbuk National University, Cheongju, South Korea
| | - Chu-Man Huang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Hui-Mei Huang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Qi-Long Cao
- Qingdao Haier Biotechnology Co., Ltd., Qingdao, China
| | - Yun-Xiao Li
- Qingdao Haier Biotechnology Co., Ltd., Qingdao, China
| | - Yong-Nan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Ying-Hua Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| |
Collapse
|
32
|
Bouyahya A, Bakrim S, Aboulaghras S, El Kadri K, Aanniz T, Khalid A, Abdalla AN, Abdallah AA, Ardianto C, Ming LC, El Omari N. Bioactive compounds from nature: Antioxidants targeting cellular transformation in response to epigenetic perturbations induced by oxidative stress. Biomed Pharmacother 2024; 174:116432. [PMID: 38520868 DOI: 10.1016/j.biopha.2024.116432] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
Oxidative stress results from a persistent imbalance in oxidation levels that promotes oxidants, playing a crucial role in the early and sustained phases of DNA damage and genomic and epigenetic instability, both of which are intricately linked to the development of tumors. The molecular pathways contributing to carcinogenesis in this context, particularly those related to double-strand and single-strand breaks in DNA, serve as indicators of DNA damage due to oxidation in cancer cases, as well as factors contributing to epigenetic instability through ectopic expressions. Oxidative stress has been considered a therapeutic target for many years, and an increasing number of studies have highlighted the promising effectiveness of natural products in cancer treatment. In this regard, we present significant research on the therapeutic targeting of oxidative stress using natural molecules and underscore the essential role of oxidative stress in cancer. The consequences of stress, especially epigenetic instability, also offer significant therapeutic prospects. In this context, the use of natural epi-drugs capable of modulating and reorganizing the epigenetic network is beginning to emerge remarkably. In this review, we emphasize the close connections between oxidative stress, epigenetic instability, and tumor transformation, while highlighting the role of natural substances as antioxidants and epi-drugs in the anti-tumoral context.
Collapse
Affiliation(s)
- Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Sara Aboulaghras
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Kawtar El Kadri
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Tarik Aanniz
- Biotechnology Lab (MedBiotech), Bioinova Research Center, Rabat Medical & Pharmacy School, Mohammed V University in Rabat, Morocco
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan PO Box: 114, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ahmed A Abdallah
- Department of Anatomy, Faculty of Medicine, Umm Alqura University, Makkah 21955, Saudi Arabia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia.
| | - Long Chiau Ming
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia; School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia; Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam.
| | - Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| |
Collapse
|
33
|
Amason ME, Li L, Harvest CK, Lacey CA, Miao EA. Validation of the Intermolecular Disulfide Bond in Caspase-2. BIOLOGY 2024; 13:49. [PMID: 38248479 PMCID: PMC10813798 DOI: 10.3390/biology13010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/05/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Caspases are a family of proteins involved in cell death. Although several caspase members have been well characterized, caspase-2 remains enigmatic. Caspase-2 has been implicated in several phenotypes, but there has been no consensus in the field about its upstream activating signals or its downstream protein targets. In addition, the unique ability of caspase-2 to form a disulfide-bonded dimer has not been studied in depth. Herein, we investigate the disulfide bond in the context of inducible dimerization, showing that disulfide bond formation is dimerization dependent. We also explore and review several stimuli published in the caspase-2 field, test ferroptosis-inducing stimuli, and study in vivo infection models. We hypothesize that the disulfide bond will ultimately prove to be essential for the evolved function of caspase-2. Proving this will require the discovery of cell death phenotypes where caspase-2 is definitively essential.
Collapse
Affiliation(s)
- Megan E. Amason
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lupeng Li
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Carissa K. Harvest
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Carolyn A. Lacey
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Edward A. Miao
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
34
|
Zhao L, Zheng L. A Review on Bioactive Anthraquinone and Derivatives as the Regulators for ROS. Molecules 2023; 28:8139. [PMID: 38138627 PMCID: PMC10745977 DOI: 10.3390/molecules28248139] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Anthraquinones are bioactive natural products, which are often found in medicinal herbs. These compounds exert antioxidant-related pharmacological actions including neuroprotective effects, anti-inflammation, anticancer, hepatoprotective effects and anti-aging, etc. Considering the benefits from their pharmacological use, recently, there was an upsurge in the development and utilization of anthraquinones as reactive oxygen species (ROS) regulators. In this review, a deep discussion was carried out on their antioxidant activities and the structure-activity relationships. The antioxidant mechanisms and the chemistry behind the antioxidant activities of both natural and synthesized compounds were furtherly explored and demonstrated. Due to the specific chemical activity of ROS, antioxidants are essential for human health. Therefore, the development of reagents that regulate the imbalance between ROS formation and elimination should be more extensive and rational, and the exploration of antioxidant mechanisms of anthraquinones may provide new therapeutic tools and ideas for various diseases mediated by ROS.
Collapse
Affiliation(s)
- Lihua Zhao
- Tianjin Renai College, Tianjin 301636, China;
| | - Lin Zheng
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|