1
|
Yang X, Luo Q, Wu Z, Wang C, Yang Y, Zheng L, Li K, Zhao L, Jurong Y. Tanshinone IIA reduces tubulointerstitial fibrosis by suppressing GSDMD-mediated pyroptosis. PHARMACEUTICAL BIOLOGY 2025; 63:364-373. [PMID: 40331369 PMCID: PMC12064128 DOI: 10.1080/13880209.2025.2498166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 03/02/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025]
Abstract
CONTEXT Tanshinone IIA (Tan IIA), a bioactive compound derived from the traditional Chinese herb Salvia miltiorrhiza (Family Lamiaceae, Authority Bunge), is well-known for its protective effects in various kidney diseases. However, its role in obstructive nephropathy has not been thoroughly investigated. OBJECTIVE This study aimed to explore the protective effects of Tan IIA in a mouse model of unilateral ureteral obstruction (UUO) and to elucidate the cellular and molecular mechanisms underlying these effects. MATERIALS AND METHODS Gasdermin D (GSDMD) knockout mice and their wild-type (WT) littermates underwent UUO surgery, with Tan IIA treatment administered 24 h prior. Human proximal tubular cells (HK-2 cells) were treated with TGF-β1 to induce fibrosis (50 ng/mL for 24 h), followed by Tan IIA treatment (5 μM) for an additional 3 h. RESULTS Tan IIA significantly reduced the expression of extracellular matrix (ECM) components, including collagen I, α-smooth muscle actin (α-SMA), vimentin and fibronectin, in UUO mice. Tan IIA attenuated GSDMD-mediated pyroptosis. However, in GSDMD knockout mice subjected to UUO, the protective effects of Tan IIA on ECM gene expression and collagen deposition in the tubular interstitium were reduced. In vitro studies showed that Tan IIA reduced GSDMD activation and fibronectin protein expression in HK-2 cells. DISCUSSION AND CONCLUSIONS Tan IIA may mitigate GSDMD-mediated pyroptosis in renal tubular epithelial cells (RTECs) and reduce kidney fibrosis, highlighting its potential as a therapeutic strategy to prevent the progression of kidney disease after ureteral obstruction.
Collapse
Affiliation(s)
- Xueling Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinglin Luo
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhifen Wu
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunxuan Wang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanjing Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Luquan Zheng
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Li
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Lei Zhao
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Jurong
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Shi J, Qin X, Sha H, Wang R, Shen H, Chen Y, Chen X. Identification of biomarkers for chronic renal fibrosis and their relationship with immune infiltration and cell death. Ren Fail 2025; 47:2449195. [PMID: 39780495 PMCID: PMC11721624 DOI: 10.1080/0886022x.2024.2449195] [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: 08/19/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) represents a significant global public health challenge. This study aims to identify biomarkers of renal fibrosis and elucidate the relationship between unilateral ureteral obstruction (UUO), immune infiltration, and cell death. METHODS Gene expression matrices for UUO were retrieved from the gene expression omnibus (GSE36496, GSE79443, GSE217650, and GSE217654). Seven genes identified through Protein-Protein Interaction (PPI) network and Support Vector Machine-Recursive Feature Elimination (SVM-RFE) analysis were validated using qRT-PCR in both in vivo and in vitro UUO experiments. WB assays were employed to investigate the role of Clec4n within NF-κB signaling pathway in renal fibrosis. The composition of immune cells in UUO was assessed using CIBERSORT, and gene set variant analysis (GSVA) was utilized to evaluate prevalent signaling pathways and cell death indices. RESULTS GO and KEGG enrichment analyses revealed numerous inflammation-related pathways significantly enriched in UUO conditions. Bcl2a1b, Clec4n, and Col1a1 were identified as potential diagnostic biomarkers for UUO. Analysis of immune cell infiltration indicated a correlation between UUO and enhanced mast cell activation. Silencing Clec4n expression appeared to mitigate the inflammatory response in renal fibrosis. GSVA results indicated elevated inflammatory pathway scores in UUO, with significant differences in disulfiram and cuproptosis scores compared to those in the normal murine kidney group. CONCLUSION Bcl2a1b, Clec4n, and Col1a1 may serve as biomarkers for diagnosing UUO. UUO development is closely linked to immune cell infiltration, activation of inflammatory pathways, disulfiram, and cuproptosis processes.
Collapse
Affiliation(s)
- Jiaqi Shi
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xinyue Qin
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Haonan Sha
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Rong Wang
- Department of Nephrology, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Hao Shen
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yinhao Chen
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| | - Xiaolan Chen
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
3
|
Yang P, Fan M, Chen Y, Yang D, Zhai L, Fu B, Zhang L, Wang Y, Ma R, Sun L. A novel strategy for the protective effect of ginsenoside Rg1 against ovarian reserve decline by the PINK1 pathway. PHARMACEUTICAL BIOLOGY 2025; 63:68-81. [PMID: 39862058 PMCID: PMC11770866 DOI: 10.1080/13880209.2025.2453699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/18/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025]
Abstract
CONTEXT The decline in ovarian reserve is a major concern in female reproductive health, often associated with oxidative stress and mitochondrial dysfunction. Although ginsenoside Rg1 is known to modulate mitophagy, its effectiveness in mitigating ovarian reserve decline remains unclear. OBJECTIVE To investigate the role of ginsenoside Rg1 in promoting mitophagy to preserve ovarian reserve. MATERIALS AND METHODS Ovarian reserve function, reproductive capacity, oxidative stress levels, and mitochondrial function were compared between ginsenoside Rg1-treated and untreated naturally aged female Drosophila using behavioral, histological, and molecular biological techniques. The protective effects of ginsenoside Rg1 were analyzed in a Drosophila model of oxidative damage induced by tert-butyl hydroperoxide. Protein expression levels in the PINK1/Parkin pathway were assessed, and molecular docking and PINK1 mutant analyses were conducted to identify potential targets. RESULTS Ginsenoside Rg1 significantly mitigated ovarian reserve decline, enhancing offspring quantity and quality, increasing the levels of ecdysteroids, preventing ovarian atrophy, and elevating germline stem cell numbers in aged Drosophila. Ginsenoside Rg1 improved superoxide dismutase, catalase activity, and gene expression while reducing reactive oxygen species levels. Ginsenoside Rg1 activated the mitophagy pathway by upregulating PINK1, Parkin, and Atg8a and downregulating Ref(2)P. Knockdown of PINK1 in the ovary by RNAi attenuated the protective effects of ginsenoside Rg1. Molecular docking analysis revealed that the ginsenoside Rg1 could bind to the active site of the PINK1 kinase domain. DISCUSSION AND CONCLUSIONS Ginsenoside Rg1 targets PINK1 to regulate mitophagy, preserving ovarian reserve. These findings suggest the potential of ginsenoside Rg1 as a therapeutic strategy to prevent ovarian reserve decline.
Collapse
Affiliation(s)
- Pengdi Yang
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Meiling Fan
- Obstetrics and Gynecology Center, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Ying Chen
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Dan Yang
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Lu Zhai
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Baoyu Fu
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Lili Zhang
- Obstetrics and Gynecology Center, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Yanping Wang
- Obstetrics and Gynecology Center, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Rui Ma
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Liwei Sun
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
4
|
Wang L, Chen SY, Li JL, Dai J, Qin DY, He RQ, Chen G. Anti-inflammatory effects of immunotherapy in clinical treatment and its potential mechanism in alleviating sleeping disorders: A systematic bibliometric study. Hum Vaccin Immunother 2025; 21:2475601. [PMID: 40097368 PMCID: PMC11917172 DOI: 10.1080/21645515.2025.2475601] [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: 10/24/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/19/2025] Open
Abstract
Sleeping disorders negatively affect cancer patient management, quality of life, and recovery. Immunotherapy, a rising cancer treatment, shows potential to improve sleep quality by reducing inflammation. This study analyzed 255 publications (2000-2024) from the Web of Science Core Collection using bibliometric methods. The US and China dominate research output, with The Mayo Clinic as a key contributor. Core topics are "immunotherapy," "quality of life," and "antibodies." Emerging keywords like "cancer," "encephalitis," and "depression" highlight a shift toward clinical psychology in treating tumors and rare diseases. It is noteworthy that with the rapid expansion of immunotherapy in cancer treatment, clinical trials have shown that it can improve sleep quality in cancer patients by reducing inflammation. As its application in cancer treatment expands, immunotherapy's potential for treating sleep disorders is promising. Future development is expected to improve sleep quality and address clinical issues, offering broad prospects for patient outcomes.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| | - Si-Yan Chen
- Day Chemotherapy Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| | - Jun-Li Li
- Day Chemotherapy Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| | - Jian Dai
- Department of Clinical Psychology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Di-Yuan Qin
- Department of Computer Science and Technology, School of Computer and Electronic Information, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| |
Collapse
|
5
|
Liu G, Kuang S, Zhong W, Yang A, Huang X, Xie Y, Zhang X, Li Y, Qin Q, Liu G. Exploring the potential mechanisms of Jinglinzi powder in treating hepatocellular carcinoma based on LC-MS, network pharmacology, molecular docking, and experimental validation. J Pharm Biomed Anal 2025; 263:116899. [PMID: 40286672 DOI: 10.1016/j.jpba.2025.116899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
This study systematically predicts the active components, targets, and mechanisms of JLZP against HCC by integrating LC-MS, network pharmacology, and molecular docking, with experimental validation of its pro-pyroptotic effects. Through HERB and NPASS databases, 81 bioactive components of JLZP and 78 overlapping HCC-related targets were identified. Protein-protein interaction network and KEGG enrichment analyses revealed that JLZP likely induces cell death via cancer-related pathways. Molecular docking (CB-Dock2) demonstrated high binding affinity between JLZP core components (e.g., protopine) and pyroptosis-associated targets (NLRP3, GSDMD). In vitro experiments confirmed that JLZP significantly suppressed MHCC-97L cell proliferation and migration, while upregulating pyroptosis markers (IL-1β, IL-18) at both mRNA and protein levels, with these effects reversed by a pyroptosis inhibitor. This study is the first to elucidate JLZP's anti-HCC mechanism through pyroptosis activation, identifying its pharmacodynamic material basis and multi-target action. The "component-target-pathway-experiment" multidimensional strategy provides methodological insights for deciphering traditional Chinese medicine formulas, offering a theoretical foundation for developing JLZP-based anticancer therapies.
Collapse
Affiliation(s)
- Gaofeng Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Shanshan Kuang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Weixing Zhong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Anming Yang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510510, China
| | - Xiaoli Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yin Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xin Zhang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510510, China
| | - Yikai Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qingguang Qin
- Department of acupuncture and moxibustion, Hainan Provincial People's Hospital, Haikou 570311, China.
| | - Guangjie Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510510, China.
| |
Collapse
|
6
|
Jian G, Wang S, Wang X, Lu Q, Zhu X, Wan S, Wang S, Li D, Wang C, He Q, Chen T, Song J. Enhanced sequential osteosarcoma therapy using a 3D-Printed bioceramic scaffold combined with 2D nanosheets via NIR-II photothermal-chemodynamic synergy. Bioact Mater 2025; 50:540-555. [PMID: 40391104 PMCID: PMC12088772 DOI: 10.1016/j.bioactmat.2025.04.029] [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: 11/08/2024] [Revised: 04/03/2025] [Accepted: 04/22/2025] [Indexed: 05/21/2025] Open
Abstract
Background Osteosarcoma (OS) is a malignant tumor originating from primitive mesenchymal cells, characterized by rapid metastasis, high invasiveness, and significant mortality. The primary challenges in OS management include the effective elimination of residual tumor cells to prevent recurrence and the repair of extensive bone defects caused by surgical intervention. Objective This study aims to develop an innovative biomimetic 3D-printed bioactive glass ceramic (BGC) scaffold modified with two-dimensional nanosheets to address both tumor ablation and bone tissue repair. Materials and methods The nanosheets were constructed via ellagic acid (EA) and ruthenium (Ru) coordination, leveraging the non-topological adhesion properties of catechol in EA to deposit the nanosheets onto the BGC scaffold (EARu-BGC). The therapeutic effects of EARu-BGC were evaluated in vitro and in vivo. Results EARu-BGC sequentially responds to the local microenvironment during OS treatment. During the tumor ablation phase, EARu-BGC induced ferroptosis through the synergistic effects of photothermal and chemodynamic therapy, achieving over 90 % tumor cell ablation and significantly inhibiting tumor volume and weight. In the bone tissue repair phase, EARu-BGC exhibited adaptive ROS scavenging and facilitated a pro-healing microenvironment, promoting osteogenic differentiation. The gradual degradation of the BGC scaffold provided essential minerals and space for new bone formation. In vivo experiments demonstrated that EARu-BGC significantly enhanced osteogenesis, increasing the trabecular number to 1.51 ± 0.15/mm and reducing trabecular separation to 1.50 ± 0.04 mm. Conclusion The EARu-BGC scaffold presents a promising multifunctional platform for OS treatment by effectively balancing antitumor efficacy with bone repair capabilities.
Collapse
Affiliation(s)
- Guangyu Jian
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Si Wang
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Xinlu Wang
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Qinyi Lu
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Xingyu Zhu
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Shucheng Wan
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Shan Wang
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Dize Li
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Chao Wang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Beihang University, Beijing 100083, PR China
| | - Qingqing He
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Tao Chen
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Jinlin Song
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| |
Collapse
|
7
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
8
|
Ji YW, Wen XY, Tang HP, Su WT, Xia ZY, Lei SQ. Necroptosis: a significant and promising target for intervention of cardiovascular disease. Biochem Pharmacol 2025; 237:116951. [PMID: 40268251 DOI: 10.1016/j.bcp.2025.116951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Due to changes in dietary structures, population aging, and the exacerbation of metabolic risk factors, the incidence of cardiovascular disease continues to rise annually, posing a significant health burden worldwide. Cell death plays a crucial role in the onset and progression of cardiovascular diseases. As a regulated endpoint encountered by cells under adverse stress conditions, the execution of necroptosis is regulated by classicalpathways, the calmodulin-dependent protein kinases (CaMK) pathway, and mitochondria-dependent pathways, and implicated in various cardiovascular diseases, including atherosclerosis, myocardial infarction, myocardial ischemia-reperfusion injury (IRI), heart failure, diabetic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, chemotherapy drug-induced cardiomyopathy, and abdominal aortic aneurysm (AAA). To further investigate potential therapeutic targets for cardiovascular diseases, we also analyzed the main molecules and their inhibitors involved in necroptosis in an effort to uncover insights for treatment.
Collapse
Affiliation(s)
- Yan-Wei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin-Yu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He-Peng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wa-Ting Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
9
|
Liu H, Wan H, Zhang A, Ouyang Y, Lu X, Wu M, Hu N, Pan J, Guo D, Li ZA, Xie D. Polypyrrole-ferric phosphate-methotrexate nanoparticles enhance apoptosis/ferroptosis of M1 macrophages via autophagy blockage for rheumatoid arthritis treatment. J Nanobiotechnology 2025; 23:428. [PMID: 40481493 PMCID: PMC12144751 DOI: 10.1186/s12951-025-03501-z] [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: 12/29/2024] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease that progresses from synovial inflammation to cartilage and bone destruction. Eliminating pro-inflammatory M1 macrophages is a promising strategy for RA treatment, but is impeded by cytoprotective autophagy. Herein, we report an effective autophagy blockage-promoted apoptosis/ferroptosis strategy using multifunctional ferric phosphate-decorated, methotrexate-loaded polypyrrole nanoparticles (PPy-FePi-MTX NPs) to achieve enhanced RA treatment effects. When injected into the knee joints of a collagen-induced DBA/1J mouse model of RA, the payloads on PPy NPs are released under the stimulation of an inflammatory microenvironment. The released MTX can directly induce M1 macrophage apoptosis. Upon near-infrared laser irradiation, the photothermal effect of PPy NPs further promotes cellular apoptosis. In addition, Fe3+ reacts with intracellular over-expressed glutathione to form Fe2+, which can convert hydrogen peroxide into toxic hydroxyl radicals. This redox process could deplete glutathione, inactivate glutathione peroxidase 4, and cause lipid peroxidation accumulation, resulting in ferroptosis of inflammatory M1 macrophages. Furthermore, PO43- disrupts the normal function of lysosomes by pH disturbance, disabling the cytoprotective autophagy of M1 macrophages for enhanced anti-RA effects. This work develops multifunctional PPy NPs for RA treatment through effective elimination of pro-inflammatory M1 macrophage.
Collapse
Affiliation(s)
- Hui Liu
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 201199, PR China
| | - Haoyu Wan
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Anbiao Zhang
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Yi Ouyang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China
| | - Xinya Lu
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Mengyuan Wu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Jianying Pan
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, PR China.
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China.
| | - Dong Guo
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, PR China.
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China.
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, NT, PR China.
| | - Denghui Xie
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, PR China.
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China.
| |
Collapse
|
10
|
Wang Y, Tang C, Wang K, Zhang X, Zhang L, Xiao X, Lin H, Xiong L. The role of ferroptosis in breast cancer: Tumor progression, immune microenvironment interactions and therapeutic interventions. Eur J Pharmacol 2025; 996:177561. [PMID: 40154567 DOI: 10.1016/j.ejphar.2025.177561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Ferroptosis represents a distinctive and distinct form of regulated cellular death, which is driven by the accumulation of lipid peroxidation. It is distinguished by altered redox lipid metabolism and is linked to a spectrum of cellular activities, including cancer. In breast cancer (BC), with triple negative breast cancer (TNBC) being an iron-and lipid-rich tumor, inducing ferroptosis was thought to be a novel approach to killing breast tumor cells. However, in the recent past, a novel conceptual framework has emerged which posits that in addition to the promotion of tumor cell death, ferritin deposition has a potent immunosuppressive effect on the tumor immune microenvironment (TIME) via the influence on both innate and adaptive immune responses. TIME of BC includes various cell populations from both the innate and adaptive immune systems. In this review, the internal association between iron homeostasis and the progression of ferroptosis, along with the common inducers and protectors of ferroptosis in BC, are discussed in detail. Furthermore, a comprehensive analysis is conducted on the dual role of ferroptosis in immune cells and proto-oncogenic functions, along with an evaluation of the potential applications of immunogenic cell death-targeted immunotherapy in TIME of BC. It is anticipated that our review will inform future research endeavors that seek to integrate ferroptosis and immunotherapy in the management of BC.
Collapse
Affiliation(s)
- Yi Wang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chuanyun Tang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Keqin Wang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaoan Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lifang Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xinghua Xiao
- Department of Pathology, The First Affiliated Hospital, Nanchang University, 17 Yongwaizheng Road, Nanschang, 330066, China
| | - Hui Lin
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lixia Xiong
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
11
|
Shao KY, Luo SD, Huang EY, Chang TM, Botcha L, Sehar M, Liu JF, Chuang PK. Acetylshikonin induces cell necroptosis via mediating mitochondrial function and oxidative stress-regulated signaling in human Oral Cancer cells. Bioorg Chem 2025; 159:108396. [PMID: 40168882 DOI: 10.1016/j.bioorg.2025.108396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025]
Abstract
Human oral squamous cell carcinoma (OSCC) represents a significant global health challenge, with conventional treatments showing limited efficacy in improving patient survival rates. To investigate the therapeutic potential of acetylshikonin on OSCC, we conducted comprehensive analyses including cell viability assays, flow cytometry, and molecular pathway investigations. Our findings demonstrate that acetylshikonin significantly inhibits OSCC cell proliferation with IC50 values of 3.81 μM and 5.87 μM in HSC3 and SCC4 cells respectively. Flow cytometry analysis revealed that acetylshikonin treatment significantly increased reactive oxygen species (ROS) production and decreased mitochondrial membrane potential in OSCC cells. Additionally, Western blot analysis showed enhanced phosphorylation of RIPK1, RIPK3, and MLKL proteins, indicating activation of the necroptotic pathway. The critical role of necroptosis was further confirmed using specific inhibitors (GSK872, Necrostatin-1, and 7-CL-O Nec-1), which significantly attenuated acetylshikonin-induced cell death. Transmission electron microscopy revealed distinct ultrastructural changes in cellular organelles, while decreased GPX4 expression suggested potential cross-activation of ferroptotic pathways. These data demonstrate that acetylshikonin suppresses OSCC growth through selective activation of oxidative stress-mediated necroptosis and mitochondrial dysfunction, identifying it as a promising natural compound for OSCC therapy through its ability to activate alternative cell death pathways and overcome traditional therapy limitations.
Collapse
Affiliation(s)
- Kung-Yu Shao
- Oral-Maxillofacial Surgery Division, Department of Dentistry, Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Sheng-Dean Luo
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Taiwan; School of Traditional Chinese Medicine, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; School of Medicine, College of Medicine, National SunYat-sen University, Kaohsiung, Taiwan
| | - Eng-Yen Huang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Taiwan; Department of Radiation Oncology & Proton and Radiation Therapy Center, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan; Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Tsung-Ming Chang
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Lavanya Botcha
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Misbah Sehar
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Po-Kai Chuang
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
12
|
Peng W, Liang J, Qian X, Li M, Nie M, Chen B. IGF2BP1/AIFM2 axis regulates ferroptosis and glycolysis to drive hepatocellular carcinoma progression. Cell Signal 2025; 130:111660. [PMID: 39971223 DOI: 10.1016/j.cellsig.2025.111660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is aggressive liver tumor that is the third leading cause of cancer death. Ferroptosis and glycolysis play key roles in HCC progression. Apoptosis-inducing factor mitochondria-associated 2 (AIFM2) in involved in regulating ferroptosis and glycolysis in cancers, but its role in HCC remains unclear. This research explored the function of AIFM2 in HCC. METHODS AIFM2 expression in HCC tissues was evaluated using the UALCAN and GEPIA databases, as well as RT-qPCR. Kaplan-Meier survival analysis analyzed the correlation between AIFM2 and the prognosis of HCC patients. EdU and transwell assays were utilized to examine HCC cell proliferation, migration, and invasion. Ferroptosis markers were analyzed by measuring iron levels, ROS production (DCFH-DA assay), and oxidative stress indicators (SOD, MDA, and GSH). Glycolytic activity was assessed through glucose uptake, lactate production, and ATP levels. m6A modification on AIFM2 mRNA was confirmed by MeRIP assay, and mRNA stability was evaluated with Actinomycin D treatment. Tumor growth and metastasis were studied in xenograft and lung metastasis models. RESULTS UALCAN analysis showed that AIFM2 was significantly upregulated in HCC tissues, which correlated with poor survival rates of HCC patients. IGF2BP1 was also highly expressed in HCC tissues and positively correlated with AIFM2 levels in HCC tissues. Functionally, AIFM2 knockdown suppressed glycolysis and enhanced ferroptosis, while its overexpression had opposite effects. IGF2BP1 was found to stabilize AIFM2 mRNA via m6A modification, promoting AIFM2 expression. IGF2BP1 knockdown reduced glycolysis, proliferation, and invasion while promoting ferroptosis, while AIFM2 overexpression could reverse this effect. In vivo, IGF2BP1 or AIFM2 silencing significantly suppressed tumor growth and metastasis. CONCLUSION IGF2BP1 stabilized AIFM2 mRNA to regulate ferroptosis and glycolysis and promoted HCC progression.
Collapse
Affiliation(s)
- Wei Peng
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Jie Liang
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Xuanlv Qian
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Mingwang Li
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Ming Nie
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Bin Chen
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China.
| |
Collapse
|
13
|
Qiu Q, Sun Q, Yang J, Yuan Q, Wang P, Liu Q, Cui Z, Ma X, Li M. The molecular mechanism by which CTSB degrades FPN to disrupt macrophage iron homeostasis and promote the progression of atherosclerosis. Mol Cell Biochem 2025; 480:3889-3906. [PMID: 39960586 DOI: 10.1007/s11010-025-05228-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/05/2025] [Indexed: 05/23/2025]
Abstract
The incidence of atherosclerosis (AS) remains high, and iron-dependent cell death (termed ferroptosis) is thought to play a key role in the progression of AS. Studies have shown that cathepsin B (CTSB) is an important regulatory molecule in atherosclerosis. However, how CTSB regulates AS progression has not been reported, and whether it is related to ferroptosis is poorly studied. In the present study, we observed a significant upregulation of CTSB expression in two AS models, ApoE knockout mice and SD rats given a HFD. According to our findings, CTSB can promote development of the AS plaque region, while inhibition of CTSB showed a reduction of AS lesion area and lipid deposition. Single-cell transcriptome analysis of AS tissue from humans revealed that CTSB is primarily expressed in macrophages. Oxidized low-density lipoprotein (ox-LDL) significantly enhanced macrophage CTSB expression, and induced ferroptosis in vitro. Mechanistically, Ferroportin (FPN) is the binding target of CTSB. CTSB can negatively regulate the protein level of FPN and promote its degradation, which further leads to ferroptosis of macrophages. We confirmed that ferroptosis in macrophages could be effectively inhibited by knockdown or pharmacological inhibition of CTSB, which in turn slowed the progression of AS. In conclusion, our study suggests that CTSB disrupts iron homeostasis in macrophages by degrading FPN and induces ferroptosis, thereby exacerbating the development of AS. Targeting CTSB may become an important potential strategy for the treatment of AS.
Collapse
Affiliation(s)
- Quanli Qiu
- Jinzhou Medical University Graduate Training Base (PLA 960, Hospital), 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Qiyu Sun
- Jinzhou Medical University Graduate Training Base (PLA 960, Hospital), 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Jiaxin Yang
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Qingxin Yuan
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Ping Wang
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Qingwei Liu
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Zhenzhen Cui
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Xiaowen Ma
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
| | - Min Li
- Jinzhou Medical University Graduate Training Base (PLA 960, Hospital), 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
| |
Collapse
|
14
|
Zhang Y, Yan Y, Liu J, Xia H, Zhou J, Cui Y, Huang X, Chang J, Zhang W, Chen W, Zhang Q, Wang S, Wang Y, Chen B. An Endoplasmic Reticulum Stress-Specific Nanoinducer Selectively Evokes Type-II Immunogenic Cell Death for Pyroptotic Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501953. [PMID: 40434207 DOI: 10.1002/adma.202501953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 05/15/2025] [Indexed: 05/29/2025]
Abstract
Specific induction of endoplasmic reticulum (ER) stress-initiated type-II immunogenic cell death (ICD) shows great potential in boosting tumor immunogenicity and anti-tumor immunotherapy. However, it remains challenging to selectively provoke type-II ICD, due to the lack of highly efficient ER targeting strategy. Here, a pH/Cathepsin-Activatable Nanoplatform (PCAN) is reported to specifically photo-induce ER stress (PCANER) and type-II ICD for cancer immunotherapy. PCANER integrates the long-circulating properties of nanomedicines with pH/cathepsin B dual-gated design, exhibiting excellent ER targeting with a colocalization efficacy of 83% in cancer tissues. Through directly intensifying glucose-regulated protein 78 and calreticulin exposure, PCANER augments type-II ICD and pyroptotic cancer cell death with high immune priming to cascade-amplify the cancer-immunity cycle, while the mild type-I ICD induced by lysosome stress (PCANLy) exhibits negligible antitumor efficacy. By leveraging the spatiotemporal subcellular organelle targeting of PCAN technology, this study achieves precise tuning of the type of ICD and cellular pyroptosis-based cancer therapy. This study offers new insights into the design of organelle level-targeted nanomedicines, paving the way for dissecting and modulating the cell death mechanism to boost cancer immunotherapy.
Collapse
Affiliation(s)
- Yimeng Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yue Yan
- Department of Central Laboratory, Peking University First Hospital, Beijing, 100034, China
| | - Jianxiong Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jiayi Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yi Cui
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xinyu Huang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jian Chang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Weiwei Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Wei Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Chemical Biology Center, Peking University, Beijing, China
- Ningbo Institute of Marine Medicine, Peking University, China
| | - Binlong Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
15
|
Zhou L, Zhao S, Xu Y, Li L, Wu Y, Zhu J, Xia D, Li F, Cai K, Zhang J. Spatial-Constraint Modulation of Intra/Extracellular Reactive Oxygen Species by Adaptive Hybrid Materials for Boosting Pyroptosis and Combined Immunotherapy of Breast Tumor. Adv Healthc Mater 2025:e2500371. [PMID: 40434189 DOI: 10.1002/adhm.202500371] [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: 01/23/2025] [Revised: 05/02/2025] [Indexed: 05/29/2025]
Abstract
Pyroptosis-immunotherapy has potential for triple-negative breast cancer treatment, but its efficacy is limited by insufficient pyroptosis activation and the need for phased, balanced, and spatially controlled activation of active species during long-term treatment. To reconcile intracellular/extracellular demands in tumor ablation, a nanoparticle-hydrogel hybrid enabling spatiotemporal reactive oxygen species (ROS) modulation is engineered. An open-shell sonosensitizer with unpaired electrons in its molecular orbitals is prepared by chelating Cu2⁺ with TCPP. These sonosensitizers are undergoing bovine serum albumin mediated biomineralization to form calcium phosphate particles and are incorporated into an injectable hydrogel through Schiff base crosslinking between dopamine-functionalized oxidized hyaluronic acid and gallic acid-modified chitosan. After intratumoral injection, nanoparticles endocytosed into tumor cells undergo acidic degradation, releasing calcium ions and GSH-activatable sonosensitizers. Calcium overload synergizes with ultrasound-mediated oxidative stress to induce mitochondrial damage and pyroptosis, while adhesive hydrogels retained in the extracellular matrix control excessive secondary ROS levels to protect oxidation-sensitive entities. This dual-action mechanism enhances the overall therapeutic effect by combining immediate tumor killing with long-term immune activation. This study provides a new route to hybrid material design, addressing the conflicting demands of short-term tumor ablation and long-term immune activation, overcoming the limitations of current pyroptosis-based immunotherapies.
Collapse
Affiliation(s)
- Luoli Zhou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Sheng Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Yijing Xu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Lin Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Yunyun Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Jing Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Daqing Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Fan Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Jixi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| |
Collapse
|
16
|
Wu Z, Wang D, Fu D, Ning D, Gu S. Rituximab-Chidamide combination chemotherapy enhances autophagy to overcome drug resistance in diffuse large B-cell lymphoma. Int Immunopharmacol 2025; 156:114578. [PMID: 40258315 DOI: 10.1016/j.intimp.2025.114578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/10/2025] [Accepted: 03/27/2025] [Indexed: 04/23/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a challenging malignancy, particularly when resistance to standard therapies such as Rituximab develops. This study investigates the combined therapeutic effects of Rituximab and Chidamide on DLBCL, focusing on drug resistance mechanisms and autophagy regulation. Using high-throughput proteomics and transcriptomic analyses, key proteins and signaling pathways were identified. BTG1 emerged as a signature gene, while autophagy-related genes such as BECN1, ATG5, HSPA8, PTEN, and MAPK8 were highlighted as pivotal players. In vitro experiments using Rituximab-sensitive and -resistant DLBCL cell lines (Raji and Raji-4RH) demonstrated that Chidamide significantly inhibited cell proliferation in a dose- and time-dependent manner, induced G0/G1 phase arrest, and enhanced autophagy. Mechanistically, Chidamide upregulated histone acetylation and autophagy-related proteins while reducing p62 levels, synergistically promoting autophagy with Rituximab. In vivo mouse models confirmed the combined treatment's efficacy in suppressing tumor growth. These findings suggest that the BTG1/BECN1/ATG5 signaling axis plays a critical role in enhancing autophagy and reversing Rituximab resistance. The combination of Chidamide and Rituximab presents a promising therapeutic strategy, offering new insights into overcoming drug resistance in DLBCL.
Collapse
Affiliation(s)
- Zelai Wu
- Department of Hematology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Dongni Wang
- Department of Hematology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Di Fu
- Department of General Practice, The Affiliated Center Hospital of Shenyang Medical College, Shenyang 110036, China
| | - Daohua Ning
- Department of Hematology, Anshan Central Hospital, Anshan 114000, China
| | - Shanshan Gu
- Department of Hematology, General Hospital of Northern Theater Command, Shenyang 110016, China.
| |
Collapse
|
17
|
Chen X, Yang M, Zhang H, Wang Y, Yan W, Cheng C, Guo R, Chai J, Zheng Y, Zhang F. Cucurbitacin B induces oral squamous cell carcinomapyroptosis via GSDME and inhibits tumour growth. Transl Oncol 2025; 58:102422. [PMID: 40424936 DOI: 10.1016/j.tranon.2025.102422] [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: 10/18/2024] [Revised: 05/12/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Pyroptosis, a form of programmed cell death, has been shown to induce anti-tumour immunity and inhibit tumour growth. Oral squamous cell carcinoma (OSCC), a prevalent malignant tumour, could benefit from pyroptosis induction as a therapeutic strategy. Cucurbitacin B (CuB), a natural compound derived from various plants, exhibits broad anti-tumour activity. However, whether CuB can exert its anti-tumour effects in OSCC through pyroptosis remains unexplored. RESULTS CuB significantly inhibited the proliferation of OSCC cells, induced pyroptosis, and elevated the levels of inflammatory factors in the cell supernatant. Bioinformatics analysis predicted the potential role of pyroptosis in OSCC, which was subsequently validated in a 4NQO-induced OSCC mouse model. The results demonstrated that CuB not only exerted tumour-inhibitory effects but also increased the infiltration of CD8+ T cells in the peritumoural region. To elucidate the mechanism of CuB-induced pyroptosis, STAT3 was identified as a key target of CuB in OSCC, with its expression upregulated in tumour tissues. Further experiments revealed that CuB induced pyroptosis by suppressing STAT3 expression and promoting the cleavage of caspase-3 and Gasdermin-E (GSDME). CONCLUSION CuB triggers OSCC pyroptosis through the STAT3/caspase-3/GSDME pathway, enhancing peritumoural CD8+ T cell infiltration and offering a novel strategy to boost tumour immunotherapy efficacy.
Collapse
Affiliation(s)
- Xin Chen
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Mengyuan Yang
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Heng Zhang
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Yajun Wang
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Wenpeng Yan
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Chen Cheng
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Rongrong Guo
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Jiawei Chai
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - YaHsin Zheng
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Fang Zhang
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China.
| |
Collapse
|
18
|
Sun X, Zhang X, Qin H, Li L. A Needle-Like H 2S-Releasing and H 2O 2 Self-Replenishing Nanoplatform for Enhanced Chemodynamic Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e06282. [PMID: 40400410 DOI: 10.1002/advs.202506282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/09/2025] [Indexed: 05/23/2025]
Abstract
The tumor microenvironment (TME) significantly restricts chemodynamic therapy (CDT) efficacy through hypoxia and antioxidant defenses. An intelligent cascade nanosystem, PTA-SnS2@GOx, is developed by integrating a tannic acid-modified Prussian blue analogue core, SnS2 shell, and glucose oxidase (GOx) activation module. The needle-like nanostructure enhanced tumor accumulation and cellular uptake. GOx-mediated glucose oxidation generated H2O2 and gluconic acid, triggering pH-responsive H2S release from SnS2. This gas disrupted mitochondrial respiration and catalase activity, alleviating hypoxia while elevating intracellular H2O2 levels. The oxygenated TME subsequently amplified GOx biocatalysis, establishing a self-sustaining cycle of H2O2 production and acidification. Concurrently, Sn4+ ions depleted glutathione, synergistically enhancing Fenton-like reactions in the PTA core for reinforced ROS generation. This multi-tiered strategy achieved effective CDT through the coordinated mechanisms: continuous H2O2 self-supply, pH reduction, and redox homeostasis disruption. Notably, the nanosystem induced immunogenic cell death, promoting dendritic cell maturation and repolarizing tumor-associated macrophages from M2 to M1 phenotype, thereby remodeling immunosuppressive TME and activating systemic antitumor immunity. The synergistic integration of self-amplifying CDT with immune sensitization demonstrates superior tumor suppression in vivo. This study provided an intelligent paradigm for cancer theranostics by combining self-supplying H2S/H2O2-enhanced CDT with sensitized immunotherapy.
Collapse
Affiliation(s)
- Xiaoxiao Sun
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Department of stomatology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xia Zhang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Haiyan Qin
- Department of stomatology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Lingling Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
19
|
Zhao G, Wang X, Wen M, Wang K, Gao X, Dou K, Yu F. Near-Infrared Fluorescence Reporter Offering Real-Time Tracking and Differential Assessment of Ferroptosis Progressions In Vivo. Anal Chem 2025. [PMID: 40402139 DOI: 10.1021/acs.analchem.5c01413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Ferroptosis-mediated tumor intervention has recently been revealed to play multiple roles in biology. Real-time monitoring of ferroptosis progression is imperative for the immediate assessment of therapeutic responses and the adjustment of diagnostic regimens but challenging as well. Herein, we present an activatable probe (DL-CL) designed for NIR fluorescence visualization and differential assessment of tumor responses to ferroptosis. DL-CL features a hemicyanine skeleton that incorporates a chlorine atom and a methylparaben group, which are quenched by a dimethylthiocarbamate-responsive group. This design enables the activation of the probe's NIR signal specifically upon exposure to the ferroptosis-related biomarker hypochlorous acid (HOCl). The rapid, highly sensitive, and specific response characteristics of DL-CL facilitate the real-time tracking of HOCl dynamics across various ferroptosis-triggering pathways in cells. Furthermore, DL-CL demonstrates its superiority in tumor identification, providing effective assistance in image-guided tumor resection and ferroptosis-mediated tumor therapy evaluation. The results reveal that increased levels of HOCl were captured following the ferroptosis combination treatment, showing a positive correlation with tumor growth inhibition. We anticipate that DL-CL may serve as a promising imaging tool for predicting the cancer therapeutic efficacy.
Collapse
Affiliation(s)
- Guiling Zhao
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Xunkai Wang
- Department of Interventional Vascular Surgery, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
| | - Muyan Wen
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Keyu Wang
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Xichen Gao
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Kun Dou
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Fabiao Yu
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
20
|
Yang C, Yu W, Dang C, Zhang J, Lu J, Xue J. FEM1B enhances TRAIL-induced apoptosis in T lymphocytes and monocytes. FEBS Open Bio 2025. [PMID: 40392678 DOI: 10.1002/2211-5463.70056] [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: 04/07/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025] Open
Abstract
FEM1B is recognized for its significant pro-apoptotic function in colorectal cancer; however, its influence and mechanisms regarding apoptosis in immune cells remain inadequately elucidated. In this study, we demonstrated that FEM1B enhances TRAIL-induced apoptosis in Molt-4, Jurkat, THP-1, and U937 cell lines. Notably, the knockdown of FEM1B in transfected cells resulted in a reversal of the observed increase in cell apoptosis. Our findings indicate that FEM1B activates caspase-3 and caspase-8, but not caspase-9, in response to TRAIL stimulation, suggesting its involvement in the extrinsic caspase-dependent apoptotic pathway. Furthermore, we found that FEM1B interacted with TRAF2 and downregulates its expression in Molt-4 and Jurkat cells, thereby diminishing TRAF2's inhibitory effect on caspase-8. In THP-1 and U937 cells, FEM1B was found to upregulate TRAIL-R2, thereby promoting TRAIL-induced apoptosis. Knockout studies in murine models further corroborated that FEM1B facilitates TRAIL-induced apoptosis. These results demonstrate that FEM1B enhances TRAIL-induced apoptosis in T lymphocytes and monocytes through a caspase-dependent mechanism involving TRAF2 or TRAIL receptors.
Collapse
Affiliation(s)
- Chenbo Yang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenhui Yu
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cui Dang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiahan Lu
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Xue
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Zeng L, Sun J, Ji K, Zhang L, Niu J, Ma K, Yan Y, Hei Z, Sun Y. CISD2 ameliorates heatstroke-associated early cognitive deficits by inhibiting ferroptosis and maintaining dendritic spine density in hippocampal neurons in mice. Neuroscience 2025; 577:282-299. [PMID: 40398723 DOI: 10.1016/j.neuroscience.2025.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/21/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
Heatstroke encephalopathy is a universal primary manifestation of heatstroke. Early brain injury caused by heatstroke may lead to long-term cognitive impairment in survivors, yet it frequently evades detection by standard diagnostic techniques. Thus, the discovery of reliable biomarkers for early brain injury detection is necessary. In this study, CISD2 downregulation in HT-22 cells was observed following hyperthermia treatment by using transcriptomics analysis. Subsequent mechanistic investigations revealed that CISD2 downregulation triggeres ferroptosis via AMPK-dependent BECN1 phosphorylation at Ser93, while CISD2 overexpression confers ferroptosis resistance in HT-22 cells at 41 °C. Furthermore, the downregulation of CISD2 expression and ferroptotic alterations were firmly observed in hippocampal tissues of mice during the early stage of heatstroke, and the overexpression of CISD2 by injecting lentivirus overexpressing CISD2 into the hippocampus of mice significantly alleviated heatstroke-induced neuronal loss, and meanwhile, the density of dendritic spines in the CA1 pyramidal neurons of the mice was effectively preserved. Moreover, knockdown of the CISD2 in the hippocampus exacerbated the loss of hippocampal neurons and the reduction of dendritic spine density. In summary, our results illustrated that CISD2 plays a pivotal role in preserving normal hippocampal function by regulating lipid peroxidation and iron homeostasis of heatstroke conditions. These finds provide novel insights into the mechanisms underlying heatstroke-induced cognitive deficits and offer potential strategies for improving risk prediction of heatstroke encephalopathy.
Collapse
Affiliation(s)
- Liang Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Jinhan Sun
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Kai Ji
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Lianxiang Zhang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, Yinchuan 750004, China
| | - Jiandong Niu
- General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Kang Ma
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Yan Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Zhiping Hei
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Yuning Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
22
|
Stachura P, Lu Z, Kronberg RM, Xu HC, Liu W, Tu JW, Schaal K, Kameri E, Picard D, von Karstedt S, Fischer U, Bhatia S, Lang PA, Borkhardt A, Pandyra AA. Deep transfer learning approach for automated cell death classification reveals novel ferroptosis-inducing agents in subsets of B-ALL. Cell Death Dis 2025; 16:396. [PMID: 40382332 DOI: 10.1038/s41419-025-07704-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
Ferroptosis is a recently described type of regulated necrotic cell death whose induction has anti-cancer therapeutic potential, especially in hematological malignancies. However, efforts to uncover novel ferroptosis-inducing therapeutics have been largely unsuccessful. In the current investigation, we classified brightfield microscopy images of tumor cells undergoing defined modes of cell death using deep transfer learning (DTL). The trained DTL network was subsequently combined with high-throughput pharmacological screening approaches using automated live cell imaging to identify novel ferroptosis-inducing functions of the polo-like kinase inhibitor volasertib. Secondary validation showed that subsets of B-cell acute lymphoblastic leukemia (B-ALL) cell lines, namely 697, NALM6, HAL01, REH and primary patient B-ALL samples were sensitive to ferroptosis induction by volasertib. This was accompanied by an upregulation of ferroptosis-related genes post-volasertib treatment in cell lines and patient samples. Importantly, using several leukemia models, we determined that volasertib delayed tumor growth and induced ferroptosis in vivo. Taken together, we have applied DTL to automated live-cell imaging in pharmacological screening to identify novel ferroptosis-inducing functions of a clinically relevant anti-cancer therapeutic.
Collapse
Affiliation(s)
- Paweł Stachura
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Zhe Lu
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Raphael M Kronberg
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
- Mathematical Modelling of Biological Systems, Heinrich Heine University, Düsseldorf, North Rhine-Westphalia, Germany
- Deep-Sea Ecology and Technology, Alfred Wegener Institute Helmholtz Centre for Polar and Marine Research, Bremerhaven, Germany
| | - Haifeng C Xu
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Wei Liu
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Jia-Wey Tu
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Katerina Schaal
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Ersen Kameri
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Cancer Prevention Graduate School (CPGS), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Silvia von Karstedt
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Weyertal 115b, Cologne, 50931, Germany
- CECAD Cluster of Excellence, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Straße 26, Cologne, 50931, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Straße 21, Cologne, 50931, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Cancer Prevention Graduate School (CPGS), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Aleksandra A Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany.
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany.
| |
Collapse
|
23
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Mechanism of ferroptosis in heart failure: The role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and intervention strategies. Ageing Res Rev 2025; 109:102770. [PMID: 40360081 DOI: 10.1016/j.arr.2025.102770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/29/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
The ferroptosis of cardiomyocytes has been recognized as the core pathological mechanism of heart failure. During the evolution of cardiovascular diseases, the accumulation of angiotensin II and advanced glycation end products can lead to the excessive activation of the RAGE/TLR4-JNK1/2 pathway, which subsequently triggers ferritinophagy, clockophagy, and enhanced p53 activity, ultimately leading to cardiomyocyte ferroptosis. It is evident that deeply unraveling the specific mechanisms in this field and comprehensively evaluating potential drugs and therapeutic strategies targeting this pathway is crucial for improving the status of cardiomyocyte ferroptosis. However, our current understanding of this pathway's specific molecular biological mechanisms in the process of cardiomyocyte ferroptosis remains limited. In light of this, this paper first comprehensively reviews the historical context of ferroptosis research, compares the similarities and differences between ferroptosis and other standard modes of cell death, elucidates the core mechanisms of ferroptosis and its close connection with heart failure, aiming to establish a basic cognitive framework for readers on ferroptosis and its role in heart failure. Subsequently, the paper delves into the pivotal role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and its intricate molecular biological regulatory network. Furthermore, it systematically integrates various therapeutic approaches aimed at inhibiting RAGE, TLR4, and JNK1/2 activity to alleviate cardiomyocyte ferroptosis, encompassing RNA interference technology, gene knockout techniques, small molecule inhibitors, natural active ingredients, as well as traditional Chinese and Western medicines, with the ultimate goal of forging new avenues and strategies for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| |
Collapse
|
24
|
Zhan X, Zheng Y, Yang J, Chen Z. Mechanism of UHRF1 protein macromolecule regulating ferroptosis in liver cancer: inhibition of GSTZ1 expression through DNA methylation. Int J Biol Macromol 2025; 311:144060. [PMID: 40348248 DOI: 10.1016/j.ijbiomac.2025.144060] [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: 03/16/2025] [Revised: 04/17/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Studies have shown that UHRF1 can affect the expression of target genes by regulating DNA methylation, which may be related to the mechanism of cancer cell death, especially ferroptosis. In this study, HuH7 and HepG2 hepatoma cell lines were used. Intracellular GSH/GSSG and NADP+/NADPH levels were assessed to analyze REDOX status, and lipid peroxidation was assessed using lipid peroxidation level assays. The regulation of GSTZ1 by UHRF1 and its modification through DNA methylation were explored. UHRF1 silencing significantly inhibited the growth and reproduction of liver cancer cells, led to iron death in HuH7 and HepG2 cells, and caused REDOX imbalance and lipid peroxidation. Further analysis showed that UHRF1 regulates iron death in liver cancer cells by mediating the expression of GSTZ1. UHRF1 can down-regulate the expression of GSTZ1 through DNA methylation modification. The study revealed that UHRF1 protein inhibits the expression of GSTZ1 by regulating DNA methylation, thereby affecting the iron death of liver cancer cells.
Collapse
Affiliation(s)
- Xiaobo Zhan
- Department of Hepatobiliary Pancreatic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| | - Yu Zheng
- Department of Hepatobiliary Pancreatic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| | - Jing Yang
- Department of Rehabilitation, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| | - Zhi Chen
- Department of Hepatobiliary Pancreatic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China.
| |
Collapse
|
25
|
Peng Y, Xu J, Wei L, Luo M, Chen S, Wei X, Luo S, Su Z, Wang Z. Melatonin alleviates sepsis-induced acute lung injury by inhibiting necroptosis via reducing circulating mtDNA release. Mol Med 2025; 31:176. [PMID: 40335920 PMCID: PMC12057123 DOI: 10.1186/s10020-025-01228-z] [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: 01/05/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Sepsis is a life-threatening condition that often leads to severe complications, including acute lung injury (ALI), which carries high morbidity and mortality in critically ill patients. Melatonin (Mel) has shown significant protective effects against sepsis-induced ALI, but its precise mechanism remains unclear. METHODS A cecal ligation and puncture (CLP) model was used to induce sepsis in male C57BL/6 mice, which were divided into four groups: Control, Sham, CLP, and CLP + Mel. ALI severity was evaluated via hematoxylin and eosin (H&E) staining, lung wet/dry ratio, and serum biomarkers (SP-D, sRAGE). Inflammatory cytokines (IL-1β, IL-6, TNF-α) were measured in serum and bronchoalveolar lavage fluid using ELISA. Circulating mitochondrial DNA (mtDNA) subtypes (D-loop, mt-CO1, mMito) were quantified by real-time PCR. TUNEL staining was performed to assess lung cell apoptosis. Necroptosis and STING pathway activation were analyzed via Western blot and immunofluorescence. RESULTS Sepsis led to increased circulating mtDNA levels and activation of necroptosis signaling pathways. Melatonin treatment alleviated sepsis-induced ALI, improving survival, reducing inflammatory cytokines and mtDNA release, and suppressing necroptosis. Intraperitoneal injection of mtDNA in mice activated necroptosis, while RIP1 inhibitor Nec-1 counteracted mtDNA-induced lung damage and necroptosis in sepsis-induced ALI. Additionally, melatonin significantly inhibited STING pathway activation. Further experiments revealed that STING modulation influenced necroptosis protein expression and mediated melatonin's protective effects in sepsis-induced ALI. CONCLUSION Melatonin mitigates sepsis-induced ALI by suppressing necroptosis through inhibition of STING activation and reduction of mtDNA release. These findings suggest melatonin as a potential therapeutic strategy for sepsis-induced ALI.
Collapse
Affiliation(s)
- Yuce Peng
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jia Xu
- Department of emergency, The first affiliated hospital of Sun Yat-sen University, Guangzhou, China
| | - Lingyu Wei
- Department of emergency, The first affiliated hospital of Sun Yat-sen University, Guangzhou, China
| | - Minghao Luo
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shenglong Chen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, China
| | - Xuebiao Wei
- Department of Geriatric Intensive Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong, China
| | - Suxin Luo
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zedazhong Su
- Department of Geriatric Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Zhonghua Wang
- Department of Geriatrics, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
26
|
Zhong C, Wang S, Zhang J, Zheng Q, Lei Y, Xu Y, Ren T, Sun R. Deoxypodophyllotoxin inhibited the growth of malignant pleural mesothelioma by inducing necroptosis and mitotic catastrophe. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156786. [PMID: 40378591 DOI: 10.1016/j.phymed.2025.156786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 04/12/2025] [Accepted: 04/17/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND Malignant pleural mesothelioma (MPM) is an extremely aggressive cancer with a poor prognosis and limited effective treatment options. However, recent studies have shown that targeting microtubule regulation is a viable approach for treating MPM. PURPOSE This study aimed to assess the antitumor behavior of deoxypodophyllotoxin (DPT) on MPM in vitro and in vivo and to identify its underlying mechanisms. STUDY DESIGN The study employed in vitro and in vivo models to evaluate the efficacy and mechanisms of DPT against MPM. We used cell-culture techniques, molecular-biology assays, and a xenograft mice module to thoroughly study the effects of DPT. METHODS Three MPM cell lines (H2452, H28, and 211H) and a xenograft mice module were used to assess the antitumor effects of DPT. The cell-cycle and cell-death rates were assessed by flow cytometry to study DPT-induced mitotic cell death. Moreover, the role of necroptosis in the antitumor effect of DPT was determined through transmission electron microscopy and western blot analysis, with further validation being done via RIP1 inhibition by Necrostatin-1 (Nec-1), a RIPK1 inhibitor, and MLKL silencing by siRNAs. RESULTS DPT was found to inhibit MPM cell growth in a dose-dependent manner in vitro and in vivo. Specifically, transmission electron microscopy showed plasma-membrane rupture with the preserved nuclear integrity of MPM cells after DPT treatment, indicating necroptosis in DPT-treated MPM cells. Moreover, a western blot revealed further proof of tumor necrosis factor alpha (TNF-α)-associated necroptosis-pathway activation, as revealed by the phosphorylation of the key proteins receptor-interacting protein kinase 1 (RIP1), receptor-interacting protein kinase 3 (RIP3), and mixed-lineage kinase domain-like pseudokinase (MLKL). Additional experiments with TNF-α receptor TNFR1 silencing, RIP1 inhibitors and MLKL silencing reinforced the influential role of TNF-α- RIP1-RIP3-MLKL activation in DPT-induced necroptosis. Also, DPT triggered mitotic catastrophe, observable by a defective spindle assembly, multinucleation, and micronucleation. Pretreatment with the S-phase arrest inducer thymidine was reduced both DPT-induced cell death and RIP1 phosphorylation, suggesting an interplay between necroptosis and mitotic arrest. CONCLUSION DPT may offer a novel therapeutic option for MPM, with drug-induced necroptosis and mitotic catastrophe being key underlying mechanisms.
Collapse
Affiliation(s)
- Cheng Zhong
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Shenqi Wang
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Jingyuan Zhang
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Qi Zheng
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yuqiong Lei
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yongle Xu
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Tao Ren
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China.
| | - Rong Sun
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China; Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Japan.
| |
Collapse
|
27
|
Shuxia Z, Ping Z, Xiaoyan Z, Sichao M, Xinyi X, Waldron K, Chenfeng W, Rouby SR, Ghonaim AH, Xingxiang C. FB1 causes barrier damage to vascular endothelial cells through ferroptosis by a PINK1/Parkin mediated mitophagy-dependent mechanism. Chem Biol Interact 2025; 416:111536. [PMID: 40324642 DOI: 10.1016/j.cbi.2025.111536] [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: 02/16/2025] [Revised: 04/19/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Fumonisin B1 (FB1) is an environmental mycotoxin produced mainly by fungi of the genus Fusarium. Exposure to FB1 can lead to pulmonary edema in pigs, likely caused by damage to vascular endothelial cells, but the mechanism of FB1-induced damage was unknown. Here, we found that FB1 damages vascular endothelial cells through ferroptosis, marked by iron-dependent membrane lipid peroxidation, and through mitophagy, a selective autophagy that targets mitochondria. FB1 exposure reduced barrier-related gene expression and increased pro-inflammatory factors. Ferroptosis was evidenced by elevated iron, ROS, lipid peroxidation, and ferroptotic markers (TFR, ACSL4), alongside decreased GSH, SLC7A11, and GPX-4 levels in vascular endothelial cells. Importantly, the ferroptosis inhibitor, Ferrostatin-1, reversed the vascular endothelial cells' barrier damage, inflammation, and ferroptosis caused by FB1. FB1-induced mitophagy was demonstrated by detecting decreased mitochondrial membrane potential and increased levels of mitophagy-related proteins. Surprisingly, silencing PINK1 using siRNA not only diminished mitophagy, cellular damage, and inflammatory responses induced by FB1, but also mitigated FB1-induced ferroptosis. In conclusion, this study demonstrates that FB1 causes vascular endothelial cell damage by ferroptosis in a mitophagy-dependent manner. This study thus lays a mechanistic foundation for the study of FB1 causing pulmonary edema in pigs and for exploring options for therapeutic intervention in conditions caused by this mycotoxin, which causes substantial harm to both human and animal health.
Collapse
Affiliation(s)
- Zhang Shuxia
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Zhang Ping
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Zheng Xiaoyan
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Mao Sichao
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Xu Xinyi
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Kevin Waldron
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Wang Chenfeng
- College of Animal Medicine, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Sherin R Rouby
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Ahmed H Ghonaim
- Department of Animal and Poultry Health, Desert Research Center, Cairo, 11435, Egypt
| | - Chen Xingxiang
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
28
|
Zheng H, Fan S, Zang H, Luo J, Shu L, Peng J. A comprehensive analysis identified an autophagy-related risk model for predicting recurrence and immunotherapy response in stage I lung adenocarcinoma. PeerJ 2025; 13:e19366. [PMID: 40330698 PMCID: PMC12051938 DOI: 10.7717/peerj.19366] [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: 11/19/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is characterized by early recurrence and poor prognosis. Autophagy is a double-edged sword in tumor development and anti-tumor therapy resistance. However, the prediction of relapse and therapeutic response in LUAD patients with stage I based on the signature of autophagy remains unclear. Methods Gene expression data were obtained from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) database. Autophagy-associated genes were extracted from the Human Autophagy Moderator Database. The autophagy score was established by Least Absolute Shrinkage and Selection Operator (LASSO) regression. Real-time PCR was used to detect gene expression of hub genes in LUAD patients. Protein-protein interaction (PPI) was analyzed to identify crucial genes. Gene set enrichment analysis (GSEA) was used to reveal the molecular features of patients. ESTIMATE algorithm was applied to estimate the tumor immune infiltration. TIDE score and Genomics of Drug Sensitivity in Cancer (GDSC) database were used to assess therapeutic response. Results We established an autophagy score based on 19 autophagy genes. Among these genes, MAP1LC3B played a crucial role in PPI network and was down-regulated in tumor tissues both in TCGA and local cohort. Receiver operating characteristic (ROC) curve showed that the risk model effectively predict RFS of stage I LUAD (area under the curve (AUC) at 1, 2, 3 years = 0.701, 0.836, and 0.818, respectively). Multivariate regression analysis indicated that the autophagy score was an independent predictor for relapse (P < 0.001, HR = 4.8, 95% CI [3.25-7.2]). The autophagy score also showed great predictive efficacy in the external validation GEO cohorts. GSEA revealed gene sets significantly enriched in immunity, cell cycle, and adhesion, etc. Meanwhile, we found the autophagy score was negatively related to KRAS mutation (P = 0.017) but positively associated with TP53 mutation (P = 6.4e-11). The autophagy score had a negative relationship with CD8+, CD4+ T cell, and dendritic cell, and positively correlated with immune checkpoint molecule CD276. Patients with a high autophagy score were sensitive to chemotherapy and targeted therapy, while resistant to immune checkpoint inhibitors. Conclusion We constructed an effective recurrence risk predictive model for stage I LUAD patients based on autophagy related genes. High autophagy score predicted a higher recurrence risk and suppressing tumor immune microenvironment.
Collapse
Affiliation(s)
- Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Long Shu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
29
|
Ouyang M, Wu J, Hu X, Liu C, Zhou D. Decoding the power of saponins in ferroptosis regulation and disease intervention: a review. J Pharm Pharmacol 2025; 77:593-608. [PMID: 39673380 DOI: 10.1093/jpp/rgae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/05/2024] [Indexed: 12/16/2024]
Abstract
OBJECTIVES This review endeavors to elucidate the complex interplay underlying diseases associated with ferroptosis and to delineate the multifaceted mechanisms by which triterpenoid and steroidal saponins modulate this form of cell death. METHODS A meticulous examination of the literature was undertaken, drawing from an array of databases including Web of Science, PubMed, and Wiley Library, with a focus on the keywords "ferroptosis," "saponin," "cancer," "inflammation," "natural products," and "signaling pathways." KEY FINDINGS Ferroptosis represents a distinctive mode of cell death that holds considerable promise for the development of innovative therapeutic strategies targeting a wide range of diseases, especially cancer and inflammatory disorders. This review reveals the nuanced interactions between saponins and critical signaling pathways, including system Xc--GSH-GPX4, Nrf2, p53, and mTOR. These interactions highlight the dual capacity of saponins to modulate ferroptosis, thereby offering fresh perspectives for therapeutic intervention. CONCLUSIONS The insights garnered from this review significantly advance our comprehension of the dynamic relationship between saponins and ferroptosis. By shedding light on these mechanisms, this work sets the stage for leveraging these insights in the creation of pioneering approaches to disease treatment, marking a significant stride in the evolution of therapeutic modalities.
Collapse
Affiliation(s)
- Min Ouyang
- College of Pharmacy, Pingxiang Health Vocational College, Pingxiang, Jiangxi, 337000, PR China
| | - Jianhua Wu
- College of Pharmacy, Pingxiang Health Vocational College, Pingxiang, Jiangxi, 337000, PR China
| | - Xizhuo Hu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Changfu Liu
- College of Pharmacy, Pingxiang Health Vocational College, Pingxiang, Jiangxi, 337000, PR China
| | - Dan Zhou
- College of Pharmacy, Pingxiang Health Vocational College, Pingxiang, Jiangxi, 337000, PR China
| |
Collapse
|
30
|
Pan R, Koo C, Su W, You Q, Guo H, Liu B. Circular RNAs modulate cell death in cardiovascular diseases. Cell Death Discov 2025; 11:214. [PMID: 40316538 PMCID: PMC12048724 DOI: 10.1038/s41420-025-02504-x] [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: 02/06/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain a global health challenge, with programmed cell death (PCD) mechanisms like apoptosis and necroptosis playing key roles in the progression. Circular RNAs (circRNAs) have recently been recognized as crucial regulators of gene expression, especially in modulating PCD. In current researches, circRNA regulation of apoptosis is the most studied area, followed by autophagy and ferroptosis. Notably, the regulatory role of circRNAs in pyroptosis and necroptosis has also begun to attract attention. From a mechanistic perspective, circRNAs influence cellular processes through several modes of action, including miRNA sponging, protein interactions, and polypeptide translation. Manipulating circRNAs and their downstream targets through inhibition or overexpression offers versatile therapeutic options for CVD treatment. Continued investigation into circRNA-mediated mechanisms may enhance our understanding of CVD pathophysiology and underscore their potential as novel and promising therapeutic targets.
Collapse
Affiliation(s)
- Runfang Pan
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chinying Koo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wenyuan Su
- Sport Medicine & Rehabilitation Center, Shanghai University of Sport, Shanghai, 200438, China
| | - Qianhui You
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haidong Guo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Baonian Liu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
31
|
Zhu Y, Yang S, Su S, Huang Y, Chen Y, Liang H, Miao J, Wu Z, Li X, Xiao J, Wang X. Macelignan Improves Functional Recovery After Spinal Cord Injury by Augmenting Autophagy via the AKT-mTOR-TFEB Signaling Pathway. Phytother Res 2025; 39:2091-2109. [PMID: 40062822 DOI: 10.1002/ptr.8473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/14/2025] [Accepted: 02/01/2025] [Indexed: 05/21/2025]
Abstract
Spinal cord injury (SCI) presents considerable therapeutic challenges due to its complex pathophysiology, and effective treatments are currently lacking. Macelignan (Mace) has shown therapeutic effects in some neurological disorders, but its potential to enhance functional recovery in SCI and the underlying mechanisms are not well understood. This research endeavors to explore the therapeutic value of Mace in SCI and its underlying mechanism of action. A mouse model of SCI was established, and the mice were randomly divided into 13 groups: Sham, Sham + Mace, SCI, SCI + 25 mg/kg Mace, SCI + Mace, SCI + 75 mg/kg Mace, SCI + 100 mg/kg Mace, SCI + 3MA, SCI + Mace/3MA, SCI + Mace/Scramble shRNA, SCI + Mace/TFEB shRNA, SCI + SC79, and SCI + Mace/SC79. Histological examinations were conducted using hematoxylin and eosin (HE), Masson's trichrome, and Nissl staining techniques. Functional recovery post-injury was evaluated through footprint analysis and the Basso Mouse Scale (BMS). The levels of proteins associated with pyroptosis and autophagy were quantified using qPCR, protein immunoblotting, and immunofluorescence (IF). Network pharmacology techniques were applied to elucidate the signaling pathways modulated by Mace. Mace facilitated functional recovery following SCI by augmenting autophagy and diminishing pyroptosis, with these effects being partially counteracted by 3-Methyladenine (3MA). It was noted that Mace induced autophagy via inhibition of the AKT-mTOR signaling pathway, leading to an increase in TFEB expression. As an autophagy activator, Mace induces TFEB-mediated autophagy and inhibits pyroptosis, which supports functional recovery post-SCI, indicating its potential clinical relevance.
Collapse
Affiliation(s)
- Yuxuan Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shu Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shenkai Su
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yeheng Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Yuli Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haibo Liang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiansen Miao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Zhejiang, China
| | - Zhouwei Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiang Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Xiao
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Zhejiang, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
32
|
Mao S, Li Q, Yang Y, Liu Z, Zhang L. Potential Crosstalk Between ANXA1+ Epithelial Cells and FABP4+ TAM Cells of Ferroptosis-Related Molecular Clusters Promotes an Immunosuppressive Microenvironment in Non-Small Cell Lung Cancer. Mol Carcinog 2025; 64:936-950. [PMID: 40040274 DOI: 10.1002/mc.23899] [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: 09/25/2024] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 03/06/2025]
Abstract
The tumor microenvironment (TME) affects tumor initiation, invasion, metastasis, and therapies. Recently, increasing evidence has demonstrated that ferroptosis plays important regulatory roles in tumourigenesis and progression. It is unclear how ferroptosis affects non-small cell lung cancer (NSCLC) progression by remodeling the TME. In this study, the single-cell RNA sequencing (scRNA-seq) data (85,562 cells, n = 18) were employed to reveal the heterogeneity of ferroptosis activation in NSCLC, and identified six ferroptosis-related molecular clusters. We found that ANXA1+ epithelial and FABP4 + TAM subpopulations were key factors in lung cancer progression and TME remodeling. In addition, the cell-cell communication analysis showed that ANXA1-FPR2/FPR1 receptor-ligand pair contributed to the formation of an immunosuppressive TME. Furthermore, we established a novel signature based on ferroptosis-related molecular clusters, and the risk score model may predict survival and response to immunotherapy. We also found that compared with responder, the expression of ANXA1 and FABP4 is higher in progressor, which indicating a higher expression of ANXA1 and FABP4 was associated with a worse response to immunotherapy. Therefore, we concluded that the molecular clusters associated with ferroptosis served as potential prognostic markers and therapeutic targets for NSCLC patients.
Collapse
Affiliation(s)
- Shengqiang Mao
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingyan Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ying Yang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhiqiang Liu
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
33
|
Kurmangaliyeva S, Baktikulova K, Tkachenko V, Seitkhanova B, Shapambayev N, Rakhimzhanova F, Almagambetova A, Kurmangaliyev K. An Overview of Hexavalent Chromium-Induced Necroptosis, Pyroptosis, and Ferroptosis. Biol Trace Elem Res 2025; 203:2619-2635. [PMID: 39287767 DOI: 10.1007/s12011-024-04376-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Heavy metals are common environmental industrial pollutants. Due to anthropogenic activity, chromium, especially its hexavalent form [Cr(VI)], is a widespread environmental contaminant that poses a threat to human health. In this review paper, we summarize the currently reported molecular mechanisms involved in chromium toxicity with a focus on the induction of pro-inflammatory non-apoptotic cell death pathways such as necroptosis, pyroptosis, and ferroptosis. The review highlights the ability of chromium to induce necroptosis, pyroptosis, and ferroptosis revealing the signaling pathways involved. Cr(VI) can induce RIPK1/RIPK3-dependent necroptosis both in vitro and in vivo. Chromium toxicity is associated with pyroptotic NLRP3 inflammasome/caspase-1/gasdermin D-dependent secretion of IL-1β and IL-18. Furthermore, this review emphasizes the role of redox imbalance and intracellular iron accumulation in Cr(VI)-induced ferroptosis. Of note, the crosstalk between the investigated lethal subroutines in chromium-induced toxicity is primarily mediated by reactive oxygen species (ROS), which are suggested to act as a rheostat determining the cell death pathway in cells exposed to chromium. The current study provides novel insights into the pro-inflammatory effects of chromium, since necroptosis, pyroptosis, and ferroptosis affect inflammation owing to their immunogenic properties linked primarily with damage-associated molecular patterns. Inhibition of these non-apoptotic lethal subroutines can be considered a therapeutic strategy to reduce the toxicity of heavy metals, including chromium.
Collapse
Affiliation(s)
- Saulesh Kurmangaliyeva
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kristina Baktikulova
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan.
| | - Viktoriya Tkachenko
- State Institution "Republican Scientific and Practical Centre of Sports, " 8 Narochanskaya St, Minsk, Republic of Belarus
| | - Bibigul Seitkhanova
- Department of Microbiology, Virology and Immunology, South Kazakhstan Medical Academy, Al-Farabi Sq, Shymkent, Republic of Kazakhstan
| | - Nasriddin Shapambayev
- Department of General Practitioner - 1, Khoja Akhmet Yasawi International Kazakh-Turkish University, 7/7 Baitursynov St, Shymkent, Republic of Kazakhstan
| | - Farida Rakhimzhanova
- Department of Microbiology, NCJSC "Semey Medical University, " 103 Abay St, Semey, Republic of Kazakhstan
| | - Altyn Almagambetova
- Department of Phthisiology and Dermatovenerology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kairat Kurmangaliyev
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| |
Collapse
|
34
|
Zhan Q, Kuang Y, Chen X, Yang Y, Jiang L, Chen J, Li L, Wang J, Zhu S, Huang H, Wang L, Zhu P, Liu R. Photo-generating Type-I ROS and aryl radicals by mitochondrial-targeting oxime-ester photogenerator for pyroptosis-mediated anti-hypoxia photoimmunotherapy. Bioact Mater 2025; 47:327-342. [PMID: 40026820 PMCID: PMC11870024 DOI: 10.1016/j.bioactmat.2025.01.032] [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: 12/15/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Pyroptosis is an inflammatory form of programmed cell death with great potential in cancer immunotherapies. Photodynamic therapy (PDT) represents a promising treatment modality to trigger pyroptosis. However, the hypoxic microenvironment inside the tumors often induces limited therapeutic efficacy. Herein, in this work, the first type of mitochondrial-targeting oxime-ester photogenerator (T-Oximer) was constructed to boost type-I ROS/aryl free radicals which could induce DNA damage by DNA cleaving and facilitate high-efficiency pyroptosis-mediated photoimmunotherapy. Detailed mechanism investigations revealed that T-Oximer could produce aryl free radicals via photolysis reaction and generate type-I ROS (O2 •- and •OH) based on the type-I electron transfer process. Meanwhile, T-Oximer could accumulate in the mitochondria, boost mitochondrial radicals, and damage mitochondria in hypoxic tumor cells. Of peculiar interest, T-Oixmer could bind with DNA and cleave DNA to induce DNA damage. Combined mitochondrial damage with DNA cleavage, T-Oximer can initiate pyroptosis, activate the ICD effect, and trigger robust systemic antitumor immunity for efficient tumor regression and metastasis suppression. Our finding provides a new strategy for constructing oxygen-independent photogenerator for high-efficiency pyroptosis-mediated anti-hypoxia photoimmunotherapy.
Collapse
Affiliation(s)
- Qiyu Zhan
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Yulin Kuang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xuyuan Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- Comprehensive Medical Treatment Ward, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanzhen Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Linhui Jiang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Jian Chen
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Lie Li
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Junwei Wang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou Avenue North No. 1838, Baiyun District, Guangzhou, 510515, China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Huanlei Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial People's Hospital Ganzhou Hospital, Ganzhou, 341000, China
| | - Lei Wang
- College of Materials and Chemical Engineering, Key Laboratory of Inorganic Nonmetallic Crystalline and Energy Conversion Materials, China Three Gorges University, Yichang, 443002, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial People's Hospital Ganzhou Hospital, Ganzhou, 341000, China
| | - Ruiyuan Liu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
35
|
Huang X, Ren X, Zhao L, Hao Y, Zhao Z, Chen F, Zhou J, Bai M, Chen S, Zhou X. Irisin Is a Potential Novel Biomarker and Therapeutic Target Against Kidney Diseases. Cell Biochem Funct 2025; 43:e70075. [PMID: 40318104 DOI: 10.1002/cbf.70075] [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: 11/04/2024] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
Kidney diseases, characterized by renal dysfunction, are the leading causes of death worldwide. It is crucial to prevent and treat kidney diseases to reduce their associated morbidity and mortality. Moderate physical exercise has been recognized to be advantageous for kidney health. Irisin is an exercise-induced myokine that was identified in 2012. It plays an important role in energy and bone metabolism, oxidative stress reduction, anti-inflammatory processes, cell death inhibition, and cardiovascular protection. However, the relationship between irisin and kidney diseases have not been fully elucidated. This review explores the role of irisin as a biomarker for kidney disease diagnosis and its associated complications, as well as the mechanisms through which it participates in various cell death pathways, such as apoptosis, autophagy, pyroptosis, and ferroptosis. Furthermore, irisin secretion levels were discussed to provide a basis for kidney disease prevention and treatment avenues, as well as therapeutic guidance for developing new and promising intervention strategies. Clinical Trial Registration: None.
Collapse
Affiliation(s)
- Xiu Huang
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Xiya Ren
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Limei Zhao
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Yajie Hao
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Zhibo Zhao
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Fahui Chen
- Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Jinxiu Zhou
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Mengqi Bai
- Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Si Chen
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Xiaoshuang Zhou
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
36
|
Nie T, Fang Y, Zhang R, Cai Y, Wang X, Jiao Y, Wu J. Self-healable and pH-responsive spermidine/ferrous ion complexed hydrogel Co-loaded with CA inhibitor and glucose oxidase for combined cancer immunotherapy through triple ferroptosis mechanism. Bioact Mater 2025; 47:51-63. [PMID: 39877156 PMCID: PMC11772096 DOI: 10.1016/j.bioactmat.2025.01.005] [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: 07/01/2024] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Tumor microenvironment governs various therapeutic tolerability of cancer such as ferroptosis and immunotherapy through rewiring tumor metabolic reprogramming like Warburg metabolism. Highly expressed carbonic anhydrases (CA) in tumor that maintaining the delicate metabolic homeostasis is thus the most potential target to be modulated to resolve the therapeutic tolerability. Hence, in this article, a self-healable and pH-responsive spermidine/ferrous ion hydrogel loaded with CA inhibitor (acetazolamide, ACZ) and glucose oxidase (ACZ/GOx@SPM-HA Gel) was fabricated through the Schiff-base reaction between spermidine-dextran and oxidized hyaluronic acid, along with ferrous coordination. Investigation on cancer cell lines (MOC-1) demonstrated ACZ/GOx@SPM-HA Gel may induce cellular oxidative stress and mitochondrial dysfunction through disrupting the cellular homeostasis. Moreover, with the facilitation of autophagy induced by spermidine, ACZ/GOx@SPM-HA Gel may trigger a positive feedback loop to maximally amplify cellular ferroptosis and promote DAMPs release. The anti-tumor evaluation on xenograft mice models furtherly proved the local injection of such hydrogel formulation could efficiently inhibit the tumor growth and distinctively promote the immunogenicity of tumor bed to provide a more favorable environment for immunotherapy. Overall, ACZ/GOx@SPM-HA Gel, with such feasible physiochemical properties and great biocompatibility, holds great potential in treating solid tumors with acidosis-mediated immunotherapy tolerance.
Collapse
Affiliation(s)
- Tianqi Nie
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
| | - Yifei Fang
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ruhe Zhang
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518106, China
| | - Yishui Cai
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, 511400, China
| | - Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518106, China
| | - Yuenong Jiao
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518106, China
| |
Collapse
|
37
|
Shan D, Song J, Ren Y, Zhang Y, Ba Y, Luo P, Cheng Q, Xu H, Weng S, Zuo A, Liu S, Han X, Deng J, Liu Z. Copper in cancer: friend or foe? Metabolism, dysregulation, and therapeutic opportunities. Cancer Commun (Lond) 2025; 45:577-607. [PMID: 39945125 PMCID: PMC12067407 DOI: 10.1002/cac2.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 05/13/2025] Open
Abstract
Copper, one of the essential nutrients for the human body, acts as an electron relay in multiple pathways due to its redox properties. Both deficiencies and excesses of copper lead to cellular fragility. Therefore, it can manifest pro- and anti-cancer properties in tumors. Therefore, it is crucial to clarify the copper activity within the cell. We have thoughtfully summarized the metabolic activities of copper from a macro and micro perspective. Cuproptosis, as well as other forms of cell death, is directly or indirectly interfered with by Cu2+, causing cancer cell death. Meanwhile, we did pan-cancer analysis of cuproptosis-related genes to further clarify the roles of these genes. In addition, copper has been found to be involved in multiple pathways within the metastasis of cancer cells. Given the complexity of copper's role, we are compelled to ask: is copper a friend or a foe? Up to now, copper has been used in various clinical applications, including protocols for measurement of copper concentration and bioimaging of radioactive 64Cu. But therapeutically it is still a continuation of the old medicine, and new possibilities need to be explored, such as the use of nanomaterials. Some studies have also shown that copper has considerable interventional power in metabolic cancers, which provides the great applications potential of copper therapy in specific cancer types. This paper reviews the dual roles played by cuproptosis in cancer from the new perspectives of oxidative stress, cell death, and tumor metastasis, and points out the value of its application in specific cancer types, summarizes the value of its testing and imaging from the perspective of clinical application as well as the current feasible options for the new use of the old drugs, and emphasizes the prospects for the application of nano-copper.
Collapse
Affiliation(s)
- Dan Shan
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
- University Hospital GalwayNational University of Ireland GalwayGalwayIreland
- Department of Biobehavioral SciencesColumbia UniversityNew YorkUSA
| | - Jinling Song
- Division of PulmonologyDepartment of PediatricsThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yuqing Ren
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Peng Luo
- The Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Quan Cheng
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanP. R. China
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanP. R. China
| | - Jinhai Deng
- Richard Dimbleby Department of Cancer ResearchComprehensive Cancer Centre, Kings College LondonLondonUK
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanP. R. China
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| |
Collapse
|
38
|
Gupta J, Mohammed MH, Alghazali T, Uthirapathy S, R R, Thakur V, Kaur M, Naidu KS, Kubaev A, Al-Mukhtar MM. Inflammasomes and autophagy in cancer: unlocking targeted therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04184-x. [PMID: 40310530 DOI: 10.1007/s00210-025-04184-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/13/2025] [Indexed: 05/02/2025]
Abstract
This study clarifies the interaction between autophagy and inflammasome within the cancer framework. The inflammasome generates pro-inflammatory cytokines to direct the immune response to pathogens and cellular stressors. Autophagy maintains cellular homeostasis and can either promote or inhibit cancer. These pathways interact to affect tumorigenesis, immune responses, and therapy. Autophagy controls inflammasome activity by affecting cancer pathogenesis and tumor microenvironment inflammation, highlighting novel cancer therapeutic approaches. Recent studies indicate that modulating autophagy and inflammasome pathways can boost anti-cancer immunity, reduce drug-resistance, and improve therapeutic efficacy. Recent studies indicate modulating inflammasome and autophagy pathways can augment anti-cancer immunity, mitigate therapy resistance, and improve treatment efficacy. Cancer research relies on understanding the inflammasome-autophagy relationship to develop targeted therapies that enhance anti-tumor efficacy and reduce inflammatory symptoms. Customized therapies may improve outcomes based on autophagy gene variations and inflammasome polymorphisms. This study investigates autophagy pathways and the inflammasome in tumor immunopathogenesis, cytokine function, and cancer therapeutic strategies, highlighting their significance in cancer biology and treatment.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India.
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-Maarif University, Anbar, Iraq.
| | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Vishal Thakur
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Manpreet Kaur
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra, Pradesh- 531162, India
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan
| | - Mahmoud Mussleh Al-Mukhtar
- Anesthesia Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
39
|
Pourhabib Mamaghani M, Mousavikia SN, Azimian H. Ferroptosis in cancer: Mechanisms, therapeutic strategies, and clinical implications. Pathol Res Pract 2025; 269:155907. [PMID: 40101548 DOI: 10.1016/j.prp.2025.155907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/10/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
The resistance of cancer cells to existing treatments has become a major challenge for researchers despite advancements in cancer treatment. Studies have shown that this resistance is due to cancer cells evading apoptosis. Moreover, the most common form of cell death induced by chemotherapy and radiotherapy is apoptosis. One of the most essential mechanisms cancer cells escape apoptosis is the excessive expression of tumors' apoptosis inhibitors. Therefore, finding a non-apoptotic pathway that bypasses apoptosis could be a hopeful strategy for cancer treatment. Ferroptosis has been identified as a non-apoptotic and regulated cell death process characterized by the accumulation of lipid peroxides and iron-dependent reactive oxygen species (ROS). Although studies have shown that ferroptosis plays a role in the development of many diseases, including cancer, it also has the potential to decrease resistance to current treatments, such as chemotherapy. Additionally, research has shown that ferroptosis successfully kills cancer cells, such as breast, stem, and lung cancer cells. Therefore, ferroptosis can be identified as a beneficial therapeutic mechanism for cancer treatment. Although ferroptosis has been introduced as an effective treatment path for cancer, its role, along with its therapeutic inducers, in increasing the therapeutic effect has not been investigated. In this review, we aim to introduce ferroptosis, compare it with other cell deaths known so far, and explain its role in cancer treatment. We believe that ferroptosis can be widely used to overcome cancer cells.
Collapse
Affiliation(s)
- Mina Pourhabib Mamaghani
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Nasibeh Mousavikia
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
40
|
Chen X, Kadier M, Shi M, Li K, Chen H, Xia Y, Wang Q, Li R, Long Y, Qin J, Wang H, Jiang G. Targeting Melatonin to Mitochondria Mitigates Castration-Resistant Prostate Cancer by Inducing Pyroptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2408996. [PMID: 40285589 DOI: 10.1002/smll.202408996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/26/2025] [Indexed: 04/29/2025]
Abstract
Prostate cancer frequently progresses to castration-resistant prostate cancer (CRPC) following androgen deprivation therapy, presenting a significant clinical challenge. Targeting tumor metabolism, particularly mitochondrial pathways, offers a promising strategy for overcoming CRPC. The modification of melatonin (Mel) to a triphenylphosphonium (TPP) cation-targeted mitochondria-melatonin (Mito-Mel) significantly increases its potency by over 1000-fold. Mito-Mel selectively targets mitochondria, enhancing reactive oxygen species (ROS) generation and causing mitochondrial membrane potential disruption. This leads to the inhibition of mitochondrial respiration including the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS), which, in turn, suppresses CRPC survival metabolic adaptations, such as glycolysis. In vitro and in vivo experiments reveal for the first time that natural small molecule compound with mitochondrial targeting via TPP exhibits excellent anticancer efficacy by inducing tumor cellular pyroptosis and facilitating the immune response, underlining the encouraging promise of this strategy for the effective treatment of CRPC.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Mairehaba Kadier
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Mengting Shi
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Kefeng Li
- Center for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macau SAR, 999078, China
| | - Hongtao Chen
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Yongzhen Xia
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Qiaohua Wang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Rongna Li
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Yili Long
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Jingbo Qin
- Guangdong Provincial Engineering Research Center of Molecular Imaging and Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine Foundation of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Hao Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| |
Collapse
|
41
|
Li T, Zhang Y, Li C, Song Y, Jiang T, Yin Y, Chang M, Song X, Zheng X, Zhang W, Yu Z, Feng W, Zhang Q, Ding L, Chen Y, Wang S. Microbial Photosynthetic Oxygenation and Radiotherapeutic Sensitization Enables Pyroptosis Induction for Combinatorial Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2503138. [PMID: 40285553 DOI: 10.1002/adma.202503138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/08/2025] [Indexed: 04/29/2025]
Abstract
Rectal cancer surgery is challenging due to the complex anatomy, making it difficult to achieve clear surgical margins. Radiotherapy (RT) plays a crucial role, especially in treating locally recurrent rectal cancer and preserving anal function. However, its effectiveness is often limited by tumor hypoxia, particularly prevalent in hypoxic regions near the bowel wall in colorectal cancer. Hypoxia contributes to both radiation resistance and apoptosis resistance, compromising RT outcomes. To overcome hypoxia-driven radiotherapy resistance, this work designs and engineers a radiotherapy-sensitizing bioplatform for efficient cancer RT. It combines lanthanum oxide nanoparticles (La2O3 NPs) with cyanobacteria, which produces oxygen through photosynthesis. This bioplatform uniquely reduces tumor hypoxia, enhances radiation deposition, and improves RT efficacy. La2O3 NPs further enhance reactive oxygen species (ROS) production induced by radiation, triggering pyroptosis via the ROS-NLRP3-GSDMD pathway, while RT amplifies pyroptosis through GSDME, circumventing tumor apoptosis resistance. The further integrated thermosensitive hydrogels ensure precise localization of the bioplatform, reducing systemic toxicity and improving therapeutic specificity. Compared to conventional therapies, this dual-action system addresses hypoxia, RT resistance, and apoptosis resistance more effectively. In vivo and in vitro hypoxia models validate its potent anti-tumor efficacy, offering valuable insights for refining clinical treatment paradigms.
Collapse
Affiliation(s)
- Tianyu Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Ya Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Cong Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Yanwei Song
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Tiaoyan Jiang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Yipengchen Yin
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xiaojun Zheng
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Wenqing Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Zhongdan Yu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Li Ding
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Sheng Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| |
Collapse
|
42
|
Jha S, Hegde M, Banerjee R, Alqahtani MS, Abbas M, Fardoun HM, Unnikrishnan J, Sethi G, Kunnumakkara AB. Nanoformulations: Reforming treatment for non-small cell lung cancer metastasis. Biochem Pharmacol 2025; 238:116928. [PMID: 40288544 DOI: 10.1016/j.bcp.2025.116928] [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: 11/21/2024] [Revised: 02/17/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) is frequently diagnosed at an advanced stage, with 20 % of cases presenting as localized disease, 25 % with regional metastasis, and 55 % with distant metastasis, contributing significantly to increased morbidity and mortality rates. Current treatments, including chemotherapy, immunotherapy, radiotherapy and targeted therapy, have shown therapeutic efficacy but are limited by issues such as lack of specificity, cytotoxicity, and therapeutic resistance. Nanoparticles (NPs) offer promising solutions to these challenges by enhancing drug penetration and retention, improving biocompatibility and stability, and achieving greater precision in targeting cancer cells. This review provides insights into various types of NPs utilized in anti-metastatic drug delivery, emphasizing their ability to enhance the efficacy of existing chemotherapeutics for the prophylaxis of metastatic NSCLC. The usage of NPs as carriers of synthetic and natural compounds aimed at inhibiting cancer cell migration and invasion have also been reviewed. Special attention has been given to biomimetic nanomaterials including extracellular vesicles and engineered exosomes, that are capable of targeting molecular pathways such as EMT, p53 and PI3K/Akt to treat metastatic NSCLC. Additionally, emphasis has been given to clinical trials of these nanoformulations and their efficacy. Although therapeutic outcomes have demonstrated certain improvements, challenges related to toxicity persist, highlighting the need for further optimization of these formulations to enhance safety and efficacy. Finally, we discuss the current limitations and future perspectives for integrating NPs into clinical settings as novel therapeutic agents for lung cancer metastasis.
Collapse
Affiliation(s)
- Shristy Jha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Mangala Hegde
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Ruchira Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Habib M Fardoun
- Research Department, Canadian University Dubai, Dubai 117781, United Arab Emirates
| | - Jyothsna Unnikrishnan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600 Singapore.
| | - Ajaikumar B Kunnumakkara
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India.
| |
Collapse
|
43
|
Song Z, Wang Q, Xiong H, Xiao J, Zhou Z, Li T, Sun Q, Qiu L, Tan Y, Liu X, Jiang H, Han S, Wang X. Bionic gene delivery system activates tumor autophagy and immunosuppressive niche to sensitize anti-PD-1 treatment against STK11-mutated lung adenocarcinoma. J Nanobiotechnology 2025; 23:312. [PMID: 40275340 PMCID: PMC12020135 DOI: 10.1186/s12951-025-03404-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
Clinical data have shown that Serine/Threonine Kinase 11 (STK11) mutation may be associated with an immunosuppressive tumor microenvironment (ITEM) and poor prognosis and failure of anti-PD-1 (αPD1) treatment in non-small cell lung cancer (NSCLC). To explore the potential of restoring STK11 protein in immunotherapy, a bionic gene delivery system was prepared by coating the STK11-encoded DNA-cationic polymer complex core with the tumor cell membrane, termed STK11@PPCM. STK11@PPCM could specifically bind with NSCLC cells and achieve precise delivery of STK11-encoded DNA. The released DNA effectively restored the STK11 protein expression, consequently reactivating autophagy and immunogenic cell death (ICD) in cancer cells. The liberated damage-associated molecular patterns (DAMPs) and autophagosome induced dendritic cells (DCs) maturation, which in turn enhanced CD8 + T cell infiltration, M1 macrophage polarization, and proinflammatory factor expression, thereby reversing the ITEM. Moreover, STK11@PPCM was also found to improve the sensitivity of cancer cells to αPD1 by increasing the expression of PD-L1, which was confirmed in STK11-mutated NSCLC cell xenografted mouse models, constructed by CRISPR-Cas9 knockout technology. This work demonstrated for the first time that restoration of functional STK11 can effectively reverse ITME and boost αPD1 efficacy in NSCLC, offering a new therapeutic approach for STK11-mutated lung adenocarcinoma in clinic.
Collapse
Affiliation(s)
- Zhongquan Song
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Qikai Wang
- Health Management Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, China
| | - Hongjie Xiong
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Jiang Xiao
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Zihan Zhou
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Tianxiang Li
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Qian Sun
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Liping Qiu
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yue Tan
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China.
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China.
| | - Shuhua Han
- Department of Pulmonary and Critical Care Medicine, Medical School, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China.
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China.
| |
Collapse
|
44
|
Ruan K, Zhang J, Chu Z, Wang X, Zhang X, Liu Q, Yang J. Exosomes in acute pancreatitis: Pathways to cellular death regulation and clinical application potential. Int Immunopharmacol 2025; 153:114491. [PMID: 40117803 DOI: 10.1016/j.intimp.2025.114491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/23/2025]
Abstract
Acute pancreatitis (AP) is a severe inflammatory condition of the digestive system which, in severe cases, can lead to persistent organ failure (POF). Developing novel therapeutic interventions and diagnostic biomarkers is critical to improve the management and prognosis of this disease. Exosomes, small extracellular vesicles, can reflect the inflammatory state of the pancreas, providing valuable insights into disease progression. Moreover, these vesicles are essential mediators of intercellular communication, modulating inflammatory responses by affecting patterns of cell death and macrophage polarization-key factors in determining AP clinical outcomes. Their stability, bioavailability, and capacity to transport various bioactive molecules render exosomes promising tools for early diagnosis and precision therapy, potentially enhancing patient outcomes. This review highlights the innovative potential of exosomes in transforming the management of AP, providing a foundation for more accurate diagnostics and targeted treatments with clinical applicability.
Collapse
Affiliation(s)
- Kaiyi Ruan
- Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Gastroenterology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Jinglei Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhuohuan Chu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang Wang
- Department of Gastroenterology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Xiaofeng Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China; Department of Gastroenterology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310006, China; Hangzhou Institute of Digestive Diseases, Hangzhou 310006, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou 310006, China.
| | - Qiang Liu
- Department of Gastroenterology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310006, China; Hangzhou Institute of Digestive Diseases, Hangzhou 310006, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou 310006, China.
| | - Jianfeng Yang
- Department of Gastroenterology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310006, China; Hangzhou Institute of Digestive Diseases, Hangzhou 310006, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou 310006, China.
| |
Collapse
|
45
|
Lie Q, Jiang H, Lu X, Chen Z, Liang J, Zhang Y, Chao H. Photo-Activated Ferrocene-Iridium(III) Prodrug Induces Immunogenic Cell Death in Melanoma Stem Cells. J Med Chem 2025; 68:8894-8906. [PMID: 40233007 DOI: 10.1021/acs.jmedchem.5c00533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cancer stem cells (CSCs) are key contributors to tumor resistance, recurrence, and metastasis. Conventional chemotherapy often fails to target and eradicate CSCs, significantly impairing their therapeutic efficacy. Herein, we design and synthesize a photoactivated ferrocene-iridium(III) complex (Ir-3) to achieve immunotherapy against melanoma cells (including stem cells). In short, Ir-3 effectively targets mitochondria and dissociates under light irradiation to produce a cytotoxic Ir(III) photosensitizer and Fe2+ ions. They can generate reactive oxygen species by the Fenton reaction, robustly induce ferroptosis and autophagy, and eventually trigger immunogenic cell death in melanoma cells (including stem cells). Furthermore, under light exposure, Ir-3 effectively inhibits stem cell-related properties and promotes macrophage-mediated phagocytosis of melanoma stem cells. For in vivo studies, Ir-3 is encapsulated in DSPE-PEG 2000 to form tumor-targeting Ir-3@PEG nanoparticles. After photoactivation, Ir-3@PEG can significantly inhibit primary and distant tumors, effectively inhibit the stemness of melanoma stem cells, and induce innate and adaptive immune responses.
Collapse
Affiliation(s)
- Qiaoshan Lie
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Hui Jiang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Xiangwan Lu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Zhuoli Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Jinzhe Liang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Yan Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
- MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 400201, P. R. China
| |
Collapse
|
46
|
Song Y, Jiang YX, Guan JY, Jiang JB, Xu MS, Zhong XY, He LN, Ren ZY, Liao Y, Liu F, Jiang YJ, Hu S, Guo W, Zhao TX, Liu XY, Gu JY, Shi YF, Luo HH, Wang K, Xiao JY. Fangchinoline-mediated autophagy inhibition amplifies antigen presentation and PD-1 blockade efficacy in lung cancer. Acta Pharmacol Sin 2025:10.1038/s41401-025-01541-7. [PMID: 40269245 DOI: 10.1038/s41401-025-01541-7] [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: 10/05/2024] [Accepted: 03/13/2025] [Indexed: 04/25/2025]
Abstract
Cancer cells frequently exhibit MHC-I deficiency, impairing immune-mediated cytotoxicity even in the presence of PD-1 checkpoint inhibition. To date, no clinically approved therapies exist that can upregulate MHC-I expression to boost immune responses against cancer cells. Emerging evidence has shown that autophagy plays a role in MHC-I molecule degradation, contributing to reduced recognition of cancer cells by CD8+ T cells. We previously report that fangchinoline, a bisbenzylisoquinoline alkaloid derived from Chinese herb, is a novel autophagy inhibitor with an adjuvant of chemotherapy against lung cancer. In this study we investigated the modulatory effects of PD-1 blockade combined with fangchinoline on CD8+ T cells within the tumor microenvironment of lung cancer. We showed an inverse correlation between elevated autophagic activity and decreased MHC-I surface expression-a phenomenon often associated with poor clinical efficacies-in various human lung cancer cell lines (NCI-H1299, NCI-H1975, A549, NCI-H1650 and NCI-H446) compared with normal bronchial epithelial cells lung cancer. Knockdown of ATG4 and ATG5 resulted in increased MHC-I expression and enhanced tumor antigen presentation in NCI-H1975, NCI-H1299 and A549 cells. As autophagy receptors were crucial for transporting proteins to autophagosomes for degradation, we sequentially silenced various autophagy receptors and found that NDP52 knockdown specifically restored MHC-I expression, suggesting that NDP52-mediated autophagy might contribute to MHC-I degradation, and autophagy inhibition might enhance immune-mediated cancer cell death. We showed that pretreatment of LLC-OVA cells with the autophagy inhibitor fangchinoline (1.25, 2.5, 5 μM) followed by coculture with CD8+ T cells, dose-dependently enhanced immune killing. In both in vitro and in vivo experiments, we showed that fangchinoline combined with anti-PD-1 therapy significantly increased CD8+ T cell-mediated cytotoxicity. In conclusion, this study highlights NDP52 as a key autophagy receptor involved in MHC-I degradation and provides a new insight into tumor immune evasion. Combining autophagy inhibition with immunotherapy may be a promising therapeutic strategy for anticancer immunity enhancement.
Collapse
Affiliation(s)
- Yue Song
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yu-Xiao Jiang
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jie-Ying Guan
- Department of Pathology and Pathophysiology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jun-Bo Jiang
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Man-Si Xu
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xue-Ying Zhong
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Li-Na He
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhen-Yang Ren
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yuan Liao
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Fang Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yan-Jun Jiang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Shan Hu
- Department of Pathology and Pathophysiology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wei Guo
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ting-Xiu Zhao
- Department of Pathology and Pathophysiology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiao-Yi Liu
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jiang-Yong Gu
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ya-Fei Shi
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huan-Huan Luo
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Kun Wang
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
- Department of Pathology and Pathophysiology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jian-Yong Xiao
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
47
|
Xu J, Weng C, Zhang Y, Zhao Q, Chen J, Pan S, Wang Y, Zhang R, Wang Y, Zhu W, Cao M, Zu D, Zhang S, Xu Z, Hu C, Cheng X. GPX4 knockdown suppresses M2 macrophage polarization in gastric cancer by modulating kynurenine metabolism. Theranostics 2025; 15:5826-5845. [PMID: 40365295 PMCID: PMC12068284 DOI: 10.7150/thno.108817] [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: 12/15/2024] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Glutathione peroxidase 4 (GPX4), an important factor regulating redox homeostasis, plays an important role in tumor microenvironment and progression. However, the role of GPX4 in gastric cancer (GC) is unclear. Methods: Spectral flow cytometry and multiplex immunohistochemistry were employed to assess the correlation between GPX4 expression and immune cell infiltration. Metabolomics analysis of conditioned media from GPX4 knockdown NUGC3 cells identified metabolic alterations. Additionally, both in vitro and in vivo functional studies were conducted to elucidate the mechanistic role of GPX4 in regulating the tumor microenvironment and progression. Results: Knockdown of GPX4 in GC cells inhibited tumor growth, enhanced CD8+ T cell infiltration, and suppressed the polarization of tumor-associated macrophages (TAMs) toward the pro-tumor M2 phenotype. Multiplex immunohistochemistry revealed a positive correlation between GPX4 expression and M2 macrophage infiltration in clinical samples from patients with GC. Metabolomics revealed that GPX4 knockdown regulate kynurenine metabolism pathway. Furthermore, mechanistic studies reveal that GPX4 silencing elevates lipid peroxidation, triggering the conversion of KYNU ubiquitin chain modifications from K48 to K63. Such ubiquitination remodeling stabilizes KYNU expression (a key kynurenine-metabolizing enzyme), reduces kynurenine accumulation, and ultimately reprograms TAM polarization to enhance antitumor immunity. We also identified that the K96 and K163 sites are important for KYNU's modification by K48 and K63 ubiquitin chains. Conclusion: Our study not only affirm the role of GPx4 in GC progression but also highlight it as a promising target for reshaping the immune microenvironment.
Collapse
Affiliation(s)
- Jingli Xu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Chunyan Weng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Qianyu Zhao
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jiahui Chen
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Siwei Pan
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yan Wang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ruolan Zhang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yuqi Wang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Weiwei Zhu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Mengxuan Cao
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Dan Zu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Shengjie Zhang
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| |
Collapse
|
48
|
Gupta G, Samuel VP, M RM, Rani B, Sasikumar Y, Nayak PP, Sudan P, Goyal K, Oliver BG, Chakraborty A, Dua K. Caspase-independent cell death in lung cancer: from mechanisms to clinical applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04149-0. [PMID: 40257494 DOI: 10.1007/s00210-025-04149-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/05/2025] [Indexed: 04/22/2025]
Abstract
Caspase-independent cell death (CICD) has recently become a very important mechanism in lung cancer, in particular, to overcome a critical failure in apoptotic cell death that is common to disease progression and treatment failures. The pathways involved in CICD span from necroptosis, ferroptosis, mitochondrial dysfunction, and autophagy-mediated cell death. Its potential therapeutic applications have been recently highlighted. Glutathione peroxidase 4 (GPX4) inhibition-driven ferroptosis has overcome drug resistance in non-small cell lung cancer (NSCLC). In addition, necroptosis involving RIPK1 and RIPK3 causes tumor cell death and modulation of immune responses in the tumor microenvironment (TME). Mitochondrial pathways are critical for CICD through modulation of metabolic and redox homeostasis. Ferroptosis is amplified by mitochondrial reactive oxygen species (ROS) and lipid peroxidation in lung cancer cells, and mitochondrial depolarization induces oxidative stress and leads to cell death. In addition, mitochondria-mediated autophagy, or mitophagy, results in the clearance of damaged organelles under stress conditions, while this function is also linked to CICD when dysregulated. The role of cell death through autophagy regulated by ATG proteins and PI3K/AKT/mTOR pathway is dual: to suppress tumor and to sensitize cells to therapy. A promising approach to enhancing therapeutic outcomes involves targeting mechanisms of CICD, including inducing ferroptosis by SLC7A11 inhibition, modulating mitochondrial ROS generation, or combining inhibition of autophagy with chemotherapy. Here, we review the molecular underpinnings of CICD, particularly on mitochondrial pathways and their potential to transform lung cancer treatment.
Collapse
Affiliation(s)
- Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vijaya Paul Samuel
- Department of Anatomy, RAK College of Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Bindu Rani
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Y Sasikumar
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Priya Priyadarshini Nayak
- Department of Medical Oncology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Puneet Sudan
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Brian G Oliver
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Amlan Chakraborty
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Kamal Dua
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia.
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
49
|
Wang S, Chen X, Wang K, Yang S. The Regulatory Role of NcRNAs in Pyroptosis and Disease Pathogenesis. Cell Biochem Biophys 2025:10.1007/s12013-025-01720-7. [PMID: 40249522 DOI: 10.1007/s12013-025-01720-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2025] [Indexed: 04/19/2025]
Abstract
Non-coding RNAs (ncRNAs), as critical regulators of gene expression, play a pivotal role in the modulation of pyroptosis and exhibit a close association with a wide range of diseases. Pyroptosis is a form of programmed cell death mediated by inflammasomes, characterized by cell membrane perforation, release of inflammatory cytokines, and a robust immune response. Recent studies have revealed that ncRNAs influence the initiation and execution of pyroptosis by regulating the expression of pyroptosis-related genes or modulating associated signaling pathways. This review systematically summarizes the molecular mechanisms and applications of ncRNAs in diseases such as cancer, infectious diseases, neurological disorders, cardiovascular diseases, and metabolic disorders. It further explores the potential of ncRNAs as diagnostic biomarkers and therapeutic targets, elucidates the intricate interactions among ncRNAs, pyroptosis, and diseases, and provides novel strategies and directions for the precision treatment of related diseases.
Collapse
Affiliation(s)
- Shaocong Wang
- Department of Cardiovascular Surgery, Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xinzhe Chen
- Department of Cardiovascular Surgery, Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Department of Cardiovascular Surgery, Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China.
| | - Sumin Yang
- Department of Cardiovascular Surgery, Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
50
|
Li J, Yu B, Xue Z, Liang Y, Chen S, Gui T, Liu Z, Zhang L, Peng R. LncRNA OLMALINC promotes osteosarcoma progression through USP1-mediated autophagy suppression. Hum Cell 2025; 38:91. [PMID: 40249458 DOI: 10.1007/s13577-025-01221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025]
Abstract
Osteosarcoma (OS) remains a challenging malignancy with poor prognosis, especially in metastatic or recurrent cases. Despite progress, the molecular mechanisms driving OS, particularly the regulation of autophagy, are not fully understood. Here, through integrated analysis of single-cell and transcriptomic data, we identify a novel long non-coding RNA (lncRNA), OLMALINC, as a critical autophagy regulator in OS. OLMALINC is significantly upregulated in OS tissues, with its expression correlating to poor clinical outcomes. Functional studies show that altering OLMALINC expression impacts OS cell progression and autophagy. Mechanistically, transcriptome analysis and RNA immunoprecipitation reveal Ubiquitin-Specific Peptidase 1 (USP1) as a direct downstream target of OLMALINC. The OLMALINC-USP1 axis inhibits autophagy and activates the hypoxia-inducible factor 1 (HIF-1α) pathway, promoting OS progression. Therapeutically, combining the USP1 inhibitor ML-323 with doxorubicin demonstrated synergistic anti-tumor effects in vitro and in vivo, enhancing autophagy and apoptosis while inhibiting tumor growth. These findings uncover a novel OLMALINC-USP1-HIF-1α axis in OS progression and highlight the potential of combining autophagy modulation with chemotherapy for improved therapeutic outcomes.
Collapse
Affiliation(s)
- Jianping Li
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bo Yu
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhaowen Xue
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yiping Liang
- Department of Basic Research Department, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Shanchuang Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Tao Gui
- Department of Orthopaedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zitao Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Lei Zhang
- Department of General Surgery, The Second Affiliated Hospital of Bengbu Medical University, Bengbu, 233080, Anhui, China.
| | - Rui Peng
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|