51
|
Leoni I, Galvani G, Monti E, Vianello C, Valenti F, Pincigher L, Grolla AA, Moro M, Coada CA, Perrone A, Righi V, Marinelli S, Ravegnini G, Giovannini C, Baldassarre M, Pariali M, Ravaioli M, Cescon M, Vasuri F, Domenicali M, Negrini M, Piscaglia F, Fato R, Stefanelli C, Gramantieri L, Bergamini C, Fornari F. MiR-22/GLUT1 Axis Induces Metabolic Reprogramming and Sorafenib Resistance in Hepatocellular Carcinoma. Int J Mol Sci 2025; 26:3808. [PMID: 40332478 PMCID: PMC12027541 DOI: 10.3390/ijms26083808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
The approval of immunotherapy has revolutionized the management of hepatocellular carcinoma (HCC) patients. However, sorafenib remains a first-line therapeutic option for advanced patients and, in particular, for patients not eligible for immune checkpoint inhibitors, but its efficacy is limited by the onset of acquired resistance, highlighting the urgent need for predictive biomarkers. This study investigates the role of miR-22 in metabolic reprogramming and its potential as a biomarker in HCC. The analysis of miR-22 expression was performed in HCC patients and preclinical models by qPCR. Functional analyses in HCC cells evaluated GLUT1 as a direct miR-22 target. Cellular and metabolic assays evaluated the miR-22/GLUT1 axis's role in metabolic changes, tumor aggressiveness, and sorafenib response. Circulating miR-22 was analyzed in sorafenib-treated HCC patients and rats. MiR-22 was downregulated in HCCs and associated with aggressive tumor features. Functionally, miR-22 modulated the HIF1A pathway, enhanced survival in stressful conditions, promoted a glycolytic shift, and enhanced cancer cell plasticity and sorafenib resistance via GLUT1 targeting. In addition, high serum miR-22 levels were associated with sorafenib resistance in HCC patients and rats. GLUT1 inhibition sensitized low miR-22-expressing HCC cells to sorafenib in preclinical models. These findings suggest that circulating miR-22 deserves attention as a predictive biomarker of sorafenib response. GLUT1 inhibition may represent a therapeutic strategy to combine with sorafenib, particularly in patients exhibiting high circulating miR-22 levels.
Collapse
Affiliation(s)
- Ilaria Leoni
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Giuseppe Galvani
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Elisa Monti
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Clara Vianello
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Francesca Valenti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Luca Pincigher
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Ambra A. Grolla
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (A.A.G.); (M.M.)
| | - Marianna Moro
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (A.A.G.); (M.M.)
| | - Camelia A. Coada
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
| | - Alessandro Perrone
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
| | - Valeria Righi
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
| | - Sara Marinelli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.M.); (L.G.)
| | - Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Catia Giovannini
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.M.); (L.G.)
| | - Maurizio Baldassarre
- Unit of Semiotics, Liver and Alcohol-Related Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Milena Pariali
- U.O. Genetica Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Matteo Ravaioli
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Hepato-Biliary Surgery and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Matteo Cescon
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Hepato-Biliary Surgery and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Vasuri
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Pathology Unit, Santa Maria delle Croci Hospital, 40121 Ravenna, Italy
| | - Marco Domenicali
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
| | - Massimo Negrini
- Department of Translational Medicine, University of Ferrara, 44100 Ferrara, Italy;
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.M.); (L.G.)
| | - Romana Fato
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Claudio Stefanelli
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
| | - Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.M.); (L.G.)
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Francesca Fornari
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
52
|
Qi Y, Tan Z, Chen H, Xiao Z, Zhang L, Wu B, Liu C, Gao Y, Yang X, Wu L, Lu L, Wang H. Pyrimidine synthase CAD deamidates and inactivates p53. Cell Res 2025:10.1038/s41422-025-01112-9. [PMID: 40240485 DOI: 10.1038/s41422-025-01112-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Affiliation(s)
- Yue Qi
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital, Fudan University, Shanghai, China
| | - Zizheng Tan
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Hanyu Chen
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital, Fudan University, Shanghai, China
| | - Ziqi Xiao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Liang Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital, Fudan University, Shanghai, China
| | - Boxuan Wu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Chennan Liu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Yunqian Gao
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xueyan Yang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital, Fudan University, Shanghai, China
| | - Lingqian Wu
- The Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics & Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Lei Lu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
| | - Hongyan Wang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Children's Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
- Prenatal Diagnosis Center of Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, Guangzhou, China.
| |
Collapse
|
53
|
Pan TT, Huang JY, Wang XD, Chen DZ, Chen YP. Copper's dual role: Reviewing its impact on liver health and disease. Int Immunopharmacol 2025; 152:114391. [PMID: 40073812 DOI: 10.1016/j.intimp.2025.114391] [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: 12/25/2024] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
As an essential trace element in the human body, Cu exists in the oxidation states of Cu(II) and Cu(I). The interconversion between these states is closely associated with various redox reactions and plays a pivotal role in cellular respiration regulation, energy metabolism, cell growth regulation, and angiogenesis promotion among other biological processes. As the primary metabolic organ, the liver synthesises and secretes Cu-binding proteins to maintain Cu homeostasis and regulate its metabolism. Studies have increasingly demonstrated that abnormally high or low levels of Cu can negatively affect the immune and metabolic microenvironment within the liver. In this review, we summarise the mechanisms underlying Cu metabolism and its dysregulation and highlight the potential involvement of disrupted Cu metabolism in several liver diseases. Our review provides insights that will help in the future development of novel therapeutic targets focusing on Cu metabolism.
Collapse
Affiliation(s)
- Tong-Tong Pan
- Hepatology Diagnosis and Treatment Center & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Jia-Yin Huang
- Hepatology Diagnosis and Treatment Center & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xiao-Dong Wang
- Hepatology Diagnosis and Treatment Center & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Da-Zhi Chen
- Hangzhou Medical College, Linan District, Hangzhou 311300, China
| | - Yong-Ping Chen
- Hepatology Diagnosis and Treatment Center & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
54
|
Xiao G, Wang X, Xu Z, Liu Y, Jing J. Lung-specific metastasis: the coevolution of tumor cells and lung microenvironment. Mol Cancer 2025; 24:118. [PMID: 40241074 PMCID: PMC12001740 DOI: 10.1186/s12943-025-02318-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
The vast majority of cancer-related deaths are attributed to metastasis. The lung, being a common site for cancer metastasis, is highly prone to being a target for multiple cancer types and causes a heavy disease burden. Accumulating evidence has demonstrated that tumor metastasis necessitates continuous interactions between tumor cells and distant metastatic niches. Nevertheless, a comprehensive elucidation of the underlying mechanisms governing lung-specific metastasis still poses a formidable challenge. In this review, we depict the lung susceptibility and the molecular profiles of tumors with the potential for lung metastasis. Under the conceptual framework of "Reciprocal Tumor-Lung Metastatic Symbiosis" (RTLMS), we mechanistically delineate the bidirectional regulatory dynamics and coevolutionary adaptation between tumor cells and distal pulmonary niches during lung-specific metastasis, including the induction of pre-metastatic-niches, positive responses of the lung, tumor colonization, dormancy, and reawakening. An enhanced understanding of the latest mechanisms is essential for developing targeted strategies to counteract lung-specific metastasis.
Collapse
Affiliation(s)
- Guixiu Xiao
- Breast Disease Center and Institute for Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xinmin Wang
- Institute of Breast Health Medicine, West China Hospital, Sichuan University Chengdu, Sichuan, 610041, China
| | - Zihan Xu
- Institute of Breast Health Medicine, West China Hospital, Sichuan University Chengdu, Sichuan, 610041, China
- Department of Medical Oncology, West China Hospital, Sichuan University, Cancer Center, Chengdu, Sichuan, 610041, China
| | - Yanyang Liu
- Department of Medical Oncology, West China Hospital, Sichuan University, Cancer Center, Chengdu, Sichuan, 610041, China.
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Jing Jing
- Breast Disease Center and Institute for Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
55
|
Bipasha M, Deepali V, Prabal D, Supriya K, Megha B. Ferroptosis: A Mechanism of Cell Death With Potential Scope in Cancer Therapy. Asia Pac J Clin Oncol 2025. [PMID: 40235436 DOI: 10.1111/ajco.14172] [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: 06/28/2024] [Revised: 11/30/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025]
Abstract
Ferroptosis is a type of regulated cell death caused by oxidative imbalance of the intracellular microenvironment. This causes the accumulation of toxic lipid peroxides, depicted by iron overload and lipid peroxidation, which results in disease development. The affected cell population displays unique morphological and biochemical features, which are distinct from other modes of cell death, like apoptosis, pyroptosis, and necroptosis. The individual pathways of each of these modes are interrelated and tend to counterbalance each other in the mechanism of cell death. The process of ferroptosis is associated with disturbances in iron metabolism, in conjunction with glutathione peroxidase and lipid peroxidation, culminating in a reduction of antioxidant capacity and accumulation of lipid peroxides in the dying cell. It has been observed that even excess cellular levels of iron can cause cell death, where ferroptosis is initiated by diminishing the levels of glutathione and glutathione peroxidase 4, and thus leading to excess build-up of lipid reactive oxygen species (ROS). In the case of a neoplastic cell, ferroptosis along with its regulators tends to orchestrate cell death and also affects cancer progression by modulation of proliferation activity, apoptosis suppression, metastasis, and drug resistance. Comprehending the complex network of molecular processes implicated in ferroptosis regulation is vital for developing targeted therapies for diseases where ferroptosis plays a significant role.
Collapse
Affiliation(s)
- Mukherjee Bipasha
- Department of Biochemistry, Dr DY Patil Medical College, Navi Mumbai, India
| | - Vidhate Deepali
- Department of Biochemistry, Dr DY Patil Medical College, Navi Mumbai, India
| | - Deb Prabal
- Sultan Qaboos Comprehensive Cancer Care & Research Centre, University Medical City, Muscat, Sultanate of Oman
| | - Khillare Supriya
- Department of Biochemistry, Dr DY Patil Medical College, Navi Mumbai, India
| | - Bangar Megha
- Department of Biochemistry, Dr DY Patil Medical College, Navi Mumbai, India
| |
Collapse
|
56
|
Kratochvilová L, Dinová A, Valková N, Dobrovolná M, Sánchez-Murcia PA, Brázda V. Chromatin Immunoprecipitation Reveals p53 Binding to G-Quadruplex DNA Sequences in Myeloid Leukemia Cell Lines. ACS BIO & MED CHEM AU 2025; 5:283-298. [PMID: 40255281 PMCID: PMC12006861 DOI: 10.1021/acsbiomedchemau.4c00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 04/22/2025]
Abstract
Clarifying functions of the p53 protein is a crucial aspect of cancer research. We analyzed the binding sites of p53 wild-type (WT) protein and its oncologically significant mutants and evaluated their transactivation properties using a functional yeast assay. Unlike the binding sites as determined in myeloid leukemia cell lines by chromatin immunoprecipitation of p53-R175H, p53-Y220C, p53-M237I, p53-R248Q, and p53-R273H mutants, the target sites of p53-WT and p53-R282W were significantly associated with putative G-quadruplex sequences (PQSs). Guanine-quadruplex (G-quadruplex or G4) formation in these sequences was evaluated by using a set of biophysical methods. G4s can modulate gene expression induced by p53. At low p53 expression level, PQS upstream of the p53-response element (RE) leads to greater gene expression induced by p53-R282W compared to that for the RE without PQS. Meanwhile, p53-WT protein expression is decreased by the PQS presence. At a high p53 expression level, the presence of PQS leads to a decreased expression of the reporter regardless of the distance and localization of the G4 from the RE.
Collapse
Affiliation(s)
- Libuše Kratochvilová
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
- Department
of Food Chemistry and Biotechnology, Faculty of Chemistry, Brno University of Technology, Purkyňova 118, Brno 612 00, Czech Republic
| | - Alessandra Dinová
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
| | - Natália Valková
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
| | - Michaela Dobrovolná
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
- Department
of Food Chemistry and Biotechnology, Faculty of Chemistry, Brno University of Technology, Purkyňova 118, Brno 612 00, Czech Republic
| | - Pedro A. Sánchez-Murcia
- Laboratory
of Computer-Aided Molecular Design, Division of Medicinal Chemistry, Otto-Loewi Research Center, Neue Stiftingtalstr. 6/III, Graz A-8010, Austria
- BioTechMed-Graz, Mozartgasse 12/II, Graz A-8010, Austria
| | - Václav Brázda
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
- Department
of Food Chemistry and Biotechnology, Faculty of Chemistry, Brno University of Technology, Purkyňova 118, Brno 612 00, Czech Republic
| |
Collapse
|
57
|
Liu J, Li Y, Ma R, Chen Y, Wang J, Zhang L, Wang B, Zhang Z, Huang L, Zhang H, Wan J, Liu H. Cold atmospheric plasma drives USP49/HDAC3 axis mediated ferroptosis as a novel therapeutic strategy in endometrial cancer via reinforcing lactylation dependent p53 expression. J Transl Med 2025; 23:442. [PMID: 40234906 PMCID: PMC11998187 DOI: 10.1186/s12967-025-06449-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Endometrial cancer ranks among the most common gynecological cancers, with increasing rates of incidence and death. Cold atmospheric plasma (CAP) has become a promising novel therapeutic approach for cancer treatment. Nevertheless, the specific impact of CAP on endometrial cancer remains inadequately characterized. OBJECTIVES This study aimed to investigate the effect of CAP on the progression of endometrial cancer and reveal its specific regulatory mechanisms. METHODS Colony formation, EdU, wound-healing, and transwell assay were used to detect the effect of CAP on endometrial cancer progression. Proteomics is employed to identify potential targets and signaling pathways through which CAP impacts endometrial cancer cells. MDA, lipid ROS, and JC-1 MMP assays were used to detect ferroptosis. Immunoprecipitation-mass spectrometry, co-immunoprecipitation, immunofluorescence co-localization, and molecular docking were used to analyze USP49 and HDAC3 interactions. The tumor xenografts model determined that CAP inhibits endometrial cancer growth in vivo. RESULTS This study observed a significant inhibitory effect of CAP on the proliferation and migration of endometrial cancer cells and reported for the first time that CAP induces ferroptosis in endometrial cancer cells. Mechanistically, CAP activated the transcription of p53 by modulating HDAC3 mediated the histone H3K18 lactylation, resulting in upregulation of p53 driving cell ferroptosis. The interaction between USP49 and HDAC3 was validated through mass spectrometry and co-immunoprecipitation experiments. The regulation of HDAC3 by CAP is contingent upon USP49, wherein the down-regulation of USP49 augments the ubiquitination of HDAC3, consequently diminishing its protein stability. Furthermore, animal models with transplanted tumors corroborated the inhibitory impact of CAP on endometrial cancer in vivo. CONCLUSIONS Our findings illustrate the suppressive effect of CAP treatment on endometrial cancer and uncover a novel regulatory mechanism mediated by CAP. Specifically, CAP modulates the ferroptosis pathway through the HDAC3/H3K18la/p53 axis, presenting a novel therapeutic approach for endometrial cancer treatment.
Collapse
Affiliation(s)
- Jinlin Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruonan Ma
- Henan Key Laboratory of Ion-beam Bioengineering, Zhengzhou University, Zhengzhou, Henan, China
| | - Youming Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jinyang Wang
- Henan Key Laboratory of Ion-beam Bioengineering, Zhengzhou University, Zhengzhou, Henan, China
| | - Lindong Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Baojin Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zidi Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lili Huang
- Department of Obstetrics and Gynecology, Yongcheng Maternal and Child Health Hospital, Shangqiu, Henan, China
| | - Hongyan Zhang
- Department of Obstetrics and Gynecology, Yongcheng Maternal and Child Health Hospital, Shangqiu, Henan, China
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
58
|
Li A, Zhao L, Liu C, Xu X, Jia J. Gray Frequency-Based Methodology for Assessing Cell Damage. ACS OMEGA 2025; 10:14084-14093. [PMID: 40256511 PMCID: PMC12004167 DOI: 10.1021/acsomega.4c11226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/22/2025]
Abstract
Cell biology techniques offer a solid foundation for evaluating and forecasting the danger of pollutants in the investigations of environmental toxicology. Studies on ecological toxicity, medication development, and illness diagnosis depend on evaluating cellular damage. The morphology of stimulated cells can alter the light scattering and reflection, and the brightness of microscopic images of the cells. This study demonstrated that stimulation-damaged and normal cells had distinct gray value distributions which led to the proposal of a novel theory to measure cellular damage by image brightness. Second, various cell types were used to confirm the method's applicability. Additionally, an evaluation technique based on gray frequency analysis can be created to determine the extent of cellular damage. This approach provides an effective and helpful tool for cellular damage visualization and quantitative evaluation in environmental toxicity assessment.
Collapse
Affiliation(s)
- Anqi Li
- Jiangmen
Key Laboratory of Synthetic Chemistry and Cleaner Production, School
of Environmental and Chemical Engineering; Carbon Neutrality Innovation
Center, Wuyi University, Jiangmen 529020, China
| | - Linying Zhao
- Jiangmen
Key Laboratory of Synthetic Chemistry and Cleaner Production, School
of Environmental and Chemical Engineering; Carbon Neutrality Innovation
Center, Wuyi University, Jiangmen 529020, China
| | - Changyu Liu
- Jiangmen
Key Laboratory of Synthetic Chemistry and Cleaner Production, School
of Environmental and Chemical Engineering; Carbon Neutrality Innovation
Center, Wuyi University, Jiangmen 529020, China
- Guangdong
Provincial Laboratory of Chemistry and Fine Chemical Industry Jieyang
Center, Jieyang 515200, China
| | - Xiaolong Xu
- Jiangmen
Key Laboratory of Synthetic Chemistry and Cleaner Production, School
of Environmental and Chemical Engineering; Carbon Neutrality Innovation
Center, Wuyi University, Jiangmen 529020, China
- Guangdong
Provincial Laboratory of Chemistry and Fine Chemical Industry Jieyang
Center, Jieyang 515200, China
| | - Jianbo Jia
- Jiangmen
Key Laboratory of Synthetic Chemistry and Cleaner Production, School
of Environmental and Chemical Engineering; Carbon Neutrality Innovation
Center, Wuyi University, Jiangmen 529020, China
- Guangdong
Provincial Laboratory of Chemistry and Fine Chemical Industry Jieyang
Center, Jieyang 515200, China
| |
Collapse
|
59
|
Huang L, Xu K, Yang Q, Ding Z, Shao Z, Li E. ANXA2 in cancer: aberrant regulation of tumour cell apoptosis and its immune interactions. Cell Death Discov 2025; 11:174. [PMID: 40234383 PMCID: PMC12000292 DOI: 10.1038/s41420-025-02469-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: 12/08/2024] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
Annexin A2 (ANXA2) is a multifunctional protein that binds to calcium and phospholipids and plays a critical role in various pathological conditions, including cancer and inflammation. Recently, there has been increasing recognition of the significant role of ANXA2 in inhibiting apoptosis and promoting immune evasion in tumour cells. Therefore, a deep understanding of the regulatory mechanisms of ANXA2 in tumour cell apoptosis and its relationship with immune evasion can provide new targets for cancer therapy. This review summarizes the role and mechanisms of ANXA2 in regulating apoptosis in tumour cells, the connection between apoptosis regulation and tumour immunity, and the potential role of ANXA2 in therapy resistance.
Collapse
Affiliation(s)
- Le Huang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Kailing Xu
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Qingping Yang
- Department of Reproductive Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai zheng Street, Nanchang, Jiangxi, 330006, China
| | - Zijun Ding
- School of Ophthalmology and Optometry, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhenduo Shao
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Enliang Li
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China.
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, Jiangxi, China.
| |
Collapse
|
60
|
Elhami A, Ghasemzadeh S, Emrahoglu S, Ghasemzadeh N, Mivehchi H, Beykoylu M, Ashrafpour M, Ayoubi S, Tabari MAK. Targeting ferroptosis: a novel pathway in oral, oropharyngeal, hypopharyngeal, and laryngeal cancers. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04142-7. [PMID: 40227310 DOI: 10.1007/s00210-025-04142-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
Malignancies of the oral cavity, oropharynx, hypopharynx, and larynx rank as the seventh most prevalent cancers globally, characterized by high morbidity and mortality. Despite advancements in conventional therapies, these cancers often demonstrate recurrence and treatment resistance. This review investigates ferroptosis, an iron-dependent regulated cell death mechanism, as a novel therapeutic target to overcome resistance and recurrence in these cancers. A narrative review study was conducted using online databases, including PubMed, Google Scholar, Scopus, and Web of Science. The search incorporated keywords such as "ferroptosis", "oral squamous cell carcinoma", "oropharyngeal cancer", "hypopharyngeal cancer", "laryngeal cancer", "iron metabolism", and "lipid peroxidation". Studies focusing on molecular mechanisms, ferroptosis regulation, and therapeutic applications were included. Key findings highlighted the involvement of genes like CA9, CAV1, and SLC7 A11 in oral squamous cell carcinoma (OSCC), contributing to progression and resistance. Ferroptosis inducers such as resveratrol and quercetin effectively promoted ferroptosis in OSCC by targeting pathways like p53/SLC7 A11. In hypopharyngeal and oropharyngeal cancers, agents like ascorbic acid and RSL3 enhanced lipid peroxidation, while laryngeal cancers showed resistance through molecules like SLC3 A2 and KPNA2, which could be counteracted with targeted therapies. Nanotechnology-based approaches, including photodynamic therapy and nanofiber membranes, offer potential for localized and effective ferroptosis induction. Ferroptosis holds promise as a therapeutic strategy for treating head and neck cancers by addressing treatment resistance and recurrence. Future research should focus on optimizing combination therapies, understanding molecular heterogeneity, and translating preclinical findings into clinical applications to improve patient outcomes.
Collapse
Affiliation(s)
- Anis Elhami
- Faculty of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | | | - Sahand Emrahoglu
- School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Nasim Ghasemzadeh
- School of Mathematics and Natural Science, University of Dallas at Texas, Richardson, TX, USA
| | - Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - Saminalsadat Ayoubi
- School of Dental Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
61
|
Wang X, Wang Z, Liu Z, Huang F, Pan Z, Zhang Z, Liu T. Nutritional strategies in oncology: The role of dietary patterns in modulating tumor progression and treatment response. Biochim Biophys Acta Rev Cancer 2025; 1880:189322. [PMID: 40228747 DOI: 10.1016/j.bbcan.2025.189322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Dietary interventions can influence tumor growth by restricting tumor-specific nutritional requirements, altering the nutrient availability in the tumor microenvironment, or enhancing the cytotoxicity of anticancer drugs. Metabolic reprogramming of tumor cells, as a significant hallmark of tumor progression, has a profound impact on immune regulation, severely hindering tumor eradication. Dietary interventions can modify tumor metabolic processes to some extent, thereby further improving the efficacy of tumor treatment. In this review, we emphasize the impact of dietary patterns on tumor progression. By exploring the metabolic differences of nutrients in normal cells versus cancer cells, we further clarify how dietary patterns influence cancer treatment. We also discuss the effects of dietary patterns on traditional treatments such as immunotherapy, chemotherapy, radiotherapy, and the gut microbiome, thereby underscoring the importance of precision nutrition.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zeyao Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zihan Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Fanxuan Huang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zhaoyu Pan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Hunan, China
| | - Zhiren Zhang
- Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China; Departments of Cardiology and Pharmacy and Breast Cancer surgery, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, China.
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China; Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.
| |
Collapse
|
62
|
Tao Y, Liu J, Lu C, Liu M, Qin X, Tian Y, Du Y. CMDF-TTS: Text-to-speech method with limited target speaker corpus. Neural Netw 2025; 188:107432. [PMID: 40249999 DOI: 10.1016/j.neunet.2025.107432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/20/2025]
Abstract
While end-to-end Text-to-Speech (TTS) methods with limited target speaker corpus can generate high-quality speech, they often require a non-target speaker corpus (auxiliary corpus) which contains a substantial amount of pairs to train the model, significantly increasing training costs. In this work, we propose a fast and high-quality speech synthesis approach, requiring few target speaker recordings. Based on statistics, we analyzed the role of phonemes, function words, and utterance target domains in the corpus and proposed a Statistical-based Compression Auxiliary Corpus algorithm (SCAC). It significantly improves model training speed without a noticeable decrease in speech naturalness. Next, we use the compressed corpus to train the proposed non-autoregressive model CMDF-TTS, which uses a multi-level prosody modeling module to obtain more information and Denoising Diffusion Probabilistic Models (DDPMs) to generate mel-spectrograms. Besides, we fine-tune the model using the target speaker corpus to embed the speaker's characteristics into the model and Conditional Variational Auto-Encoder Generative Adversarial Networks(CVAE-GAN) to enhance further the synthesized speech's quality. Experimental results on multiple Mandarin and English corpus demonstrate that the CMDF-TTS model, enhanced by the SCAC algorithm, effectively balances training speed and synthesized speech quality. Overall, its performance surpasses that of state-of-the-art models.
Collapse
Affiliation(s)
- Ye Tao
- School of Information Science and Technology, Qingdao University of Science and Technology, Qingdao 266061, PR China.
| | - Jiawang Liu
- School of Information Science and Technology, Qingdao University of Science and Technology, Qingdao 266061, PR China.
| | - Chaofeng Lu
- School of Information Science and Technology, Qingdao University of Science and Technology, Qingdao 266061, PR China.
| | - Meng Liu
- School of Reliability and Systems Engineering, Beihang University, Beijing 100191, PR China.
| | - Xiugong Qin
- Beijing Research Institute of Automation for Machinery Industry Co., Ltd., Beijing 100000, PR China.
| | - Yunlong Tian
- National Engineering Research Center of Digital Home Networking, Qingdao 266000, PR China.
| | - Yongjie Du
- National Engineering Research Center of Digital Home Networking, Qingdao 266000, PR China.
| |
Collapse
|
63
|
Ren W, Yu Y, Wang T, Wang X, Su K, Wang Y, Tang W, Liu M, Zhang Y, Yang L, Diao H. Comprehensive analysis of metabolism-related gene biomarkers reveals their impact on the diagnosis and prognosis of triple-negative breast cancer. BMC Cancer 2025; 25:668. [PMID: 40217479 PMCID: PMC11987350 DOI: 10.1186/s12885-025-14053-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by poor prognosis and limited treatment options, which underscores the urgency of the discovery of new biomarkers. Metabolic reprogramming is a hallmark of cancer and is expected to serve as a strong predictive biomarker for breast cancer. METHODS We integrated RNA expression data and clinical information from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases to explore the associations between metabolism-related gene expression and TNBC prognosis. Our comprehensive approach included differential expression analysis, enrichment analysis, Cox regression analysis, machine learning, and in vitro experimental validation. RESULTS We identified five pivotal genes-SDS, RDH12, IDO1, GLDC, and ALOX12B-that were significantly correlated with the prognosis of TNBC patients. A prognostic model incorporating these genes was developed and validated in an independent patient cohort. The model demonstrated predictive validity, as TNBC patients classified into the high-risk group exhibited significantly poorer prognoses. Additionally, utilizing the risk model, we evaluated the mutational landscape, immune infiltration, immunotherapy response, and drug sensitivity in TNBC, providing insights into potential therapeutic strategies. CONCLUSIONS This study established a robust prognostic model capable of accurately predicting clinical outcomes and metastasis, which could aid in personalized clinical decision-making.
Collapse
Affiliation(s)
- Weibin Ren
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
| | - Yuyun Yu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
| | - Xueyao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
| | - Kunkai Su
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanbo Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
| | - Wenjie Tang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
| | - Miaomiao Liu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
| | - Yanhui Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
| | - Long Yang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China
| | - Hongyan Diao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, Shandong, China.
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
64
|
Cao Y, He Y, Liao L, Xu L. Circular RNAs perspective: exploring the direction of immunotherapy for colorectal cancer. Front Oncol 2025; 15:1554179. [PMID: 40291917 PMCID: PMC12021614 DOI: 10.3389/fonc.2025.1554179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Circular RNAs (circRNAs) are multifaceted molecules that play a pivotal role in regulating gene expression at both transcriptional and post-transcriptional levels. Their expression is highly tissue-specific and developmentally regulated, making them critical players in various physiological processes and diseases, particularly cancer. In colorectal cancer, circRNAs exhibit significantly dysregulated expression patterns and profoundly influence disease progression through diverse molecular mechanisms. Unraveling the complex roles of circRNAs in modulating colorectal cancer immunotherapy outcomes highlights their potential as both promising biomarkers and therapeutic targets. Moving forward, advancements in circRNA-based therapeutic strategies and delivery systems are poised to transform precision medicine, enabling early colorectal cancer diagnosis and improving patient prognosis.
Collapse
Affiliation(s)
- Yanlin Cao
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
- The First Clinical Medical College of Guangdong Medical University, Zhanjiang, China
| | - Yuxing He
- Department of Medical Laboratory Medicine, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
| | - Lingshan Liao
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
| | - Lixin Xu
- Neurosurgery Department, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
| |
Collapse
|
65
|
Gnanamony M, Thomas M, Nguyen TH, Brownstein K, de Alarcon PA. Pomiferin Induces Antiproliferative and Pro-Death Effects in High-Risk Neuroblastoma Cells by Modulating Multiple Cell Death Pathways. Int J Mol Sci 2025; 26:3600. [PMID: 40332068 PMCID: PMC12026727 DOI: 10.3390/ijms26083600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Resistance to chemotherapy-induced apoptosis significantly hinders the successful treatment of high-risk neuroblastoma (NB). Natural compounds, such as osajin and pomiferin-isoflavones extracted from Osage orange (Maclura pomifera [Raf.] Schneid.)-have known anti-inflammatory and anticancer properties and may have the potential as a therapeutic agent to target conventional drug resistance in NB. In this study, we investigated the antiproliferative and cytotoxic potential of osajin and pomiferin in NB cell lines. Both compounds reduced proliferation and induced cytotoxicity, with pomiferin showing a lower IC50 than osajin. Using multiple techniques, we show that pomiferin induced a dose-dependent increase in apoptosis. In addition to apoptosis, we identified the activation of multiple cell death pathways. Pomiferin induced ferroptosis by inhibiting GPX4 and increasing lipid peroxidation. In addition, pomiferin treatment significantly impaired autophagic machinery. LAN5, a MYCN-amplified cell line, showed increased gasdermin E cleavage in response to pomiferin, suggesting pyroptosis. No changes were observed in phosphorylated MLKL, indicating the absence of necroptosis. In conclusion, our comprehensive evaluation demonstrates that pomiferin activates multiple cell death pathways in high-risk NB cells, potentially offering a valuable strategy to overcome drug resistance to conventional chemotherapy.
Collapse
Affiliation(s)
- Manu Gnanamony
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (P.A.d.A.)
| | - Maria Thomas
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (P.A.d.A.)
| | - Thu Hien Nguyen
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (P.A.d.A.)
| | - Korey Brownstein
- Functional Foods Research Unit, United States Department of Agriculture, Agricultural Research Service, National Center for Agricultural Utilization Research, Peoria, IL 61604, USA;
| | - Pedro A. de Alarcon
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (P.A.d.A.)
| |
Collapse
|
66
|
Borkosky SS, Peralta-Martínez R, Armella-Sierra A, Esperante SA, Lizárraga L, García-Pardo J, Ventura S, Sánchez IE, de Prat-Gay G. Experimental kinetic mechanism of P53 condensation-amyloid aggregation. Biophys J 2025:S0006-3495(25)00236-X. [PMID: 40221836 DOI: 10.1016/j.bpj.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/03/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025] Open
Abstract
The tumor suppressor p53 modulates the transcription of a variety of genes, constituting a protective barrier against anomalous cellular proliferation. High-frequency "hotspot" mutations result in loss of function by the formation of amyloid-like aggregates that correlate with cancerous progression. We show that full-length p53 undergoes spontaneous homotypic condensation at submicromolar concentrations and in the absence of crowders to yield dynamic coacervates that are stoichiometrically dissolved by DNA. These coacervates fuse and evolve into hydrogel-like clusters with strong thioflavin T binding capacity, which further evolve into fibrillar species with a clearcut branching growth pattern. The amyloid-like coacervates can be rescued by the human papillomavirus master regulator E2 protein to yield large regular droplets. Furthermore, we kinetically dissected an overall condensation mechanism, which consists of a nucleation-growth process by the sequential addition of p53 tetramers, leading to discretely sized and monodisperse early condensates followed by coalescence into bead-like coacervates that slowly evolve to the fibrillar species. Our results suggest strong similarities to condensation-to-amyloid transitions observed in neurological aggregopathies. Mechanistic insights uncover novel key early and intermediate stages of condensation that can be targeted for p53 rescuing drug discovery.
Collapse
Affiliation(s)
- Silvia S Borkosky
- Laboratorio de Estructura-Función e Ingeniería de Proteínas, Fundación Instituto Leloir- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIB-BA), Buenos Aires, Argentina
| | - Ramón Peralta-Martínez
- Laboratorio de Estructura-Función e Ingeniería de Proteínas, Fundación Instituto Leloir- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIB-BA), Buenos Aires, Argentina
| | - Alicia Armella-Sierra
- Laboratorio de Estructura-Función e Ingeniería de Proteínas, Fundación Instituto Leloir- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIB-BA), Buenos Aires, Argentina
| | - Sebastián A Esperante
- Centro de Rediseño de Proteínas (CRIP), CONICET, 25 de Mayo y Francia (1650), Universidad Nacional de San Martin (UNSAM), Buenos Aires, Argentina
| | - Leonardo Lizárraga
- Centro de Investigaciones en Bionanociencias (CIBION), Buenos Aires, Argentina
| | - Javier García-Pardo
- Institut de Biotecnologia i de Biomedicina (IBB) and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i de Biomedicina (IBB) and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ignacio E Sánchez
- Laboratorio de Fisiología de Proteínas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gonzalo de Prat-Gay
- Laboratorio de Estructura-Función e Ingeniería de Proteínas, Fundación Instituto Leloir- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIB-BA), Buenos Aires, Argentina.
| |
Collapse
|
67
|
Wang X, Xue Y, Chang L, Zhu X, Liu W, Liang T. The Regulation of Trace Metal Elements in Cancer Ferroptosis. Adv Biol (Weinh) 2025:e2400821. [PMID: 40200790 DOI: 10.1002/adbi.202400821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/11/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, as novel type of regulated cell death that has garnered widespread attention over the past decade, has witnessed the continuous discovery of an increasing number of regulatory mechanisms. Trace metal elements play a multifaceted and crucial role in oncology. Interestingly, it has been increasingly evident that these elements, such as copper, are involved in the regulation of iron accumulation, lipid peroxidation and antiferroptotic systems, suggesting the existence of "nonferrous" mechanisms in ferroptosis. In this review, a comprehensive overview of the composition and mechanism of ferroptosis is provided. The interaction between copper metabolism (including cuproptosis) and ferroptosis in cancer, as well as the roles of other trace metal elements (such as zinc, manganese, cobalt, and molybdenum) in ferroptosis are specifically focused. Furthermore, the applications of nanomaterials based on these metals in cancer therapy are also reviewed and potential strategies for co-targeting ferroptosis and cuproptosis are explored. Nevertheless, in light of the intricate and ambiguous nature of these interactions, ongoing research is essential to further elucidate the "nonferrous" mechanisms of ferroptosis, thereby facilitating the development of novel therapeutic targets and approaches for cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuanyuan Xue
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lei Chang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xuena Zhu
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Wenjun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
- Zhejiang University Cancer Center, Hangzhou, 310003, China
| |
Collapse
|
68
|
Chen S, Hao Q, Gan Y, Tong J, Xiong C, Liao Q, Zhang Y, Ye T, Zhou X, Chen H. p53 transcriptionally activates DCP1B to suppress tumor progression and enhance tumor sensitivity to PI3K blockade in non-small cell lung cancer. Cell Death Differ 2025:10.1038/s41418-025-01501-y. [PMID: 40200093 DOI: 10.1038/s41418-025-01501-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
Non-small cell lung cancer (NSCLC), which accounts for approximately 85% of lung cancer patients, is characterized by its aggressive nature and poor prognosis. In this study, we identify decapping mRNA 1B (DCP1B) as a tumor suppressor gene that is transcriptionally regulated by p53. DCP1B is found to inhibit the growth and migration of NSCLC cells. Consistently, the level of DCP1B expression is decreased in NSCLC tissues, and its low expression is associated with NSCLC patients' unfavorable outcomes. Mechanistic investigations reveal that DCP1B promotes the turnover of mitogen-activated protein kinase 4 (MAPK4) mRNA, and the activation of p53 reduces the expression level of MAPK4 partially through DCP1B. Notably, overexpression of MAPK4 can drive AKT phosphorylation independent of phosphoinositide 3-kinase (PI3K), thus neutralizing the anti-tumor activity of the PI3K inhibitor in NSCLC cells. Moreover, the p53 agonist combined with the PI3K inhibitor can suppress NSCLC proliferation synergistically in vitro and in vivo. Collectively, this study not only uncovers the function and mechanism of the p53-DCP1B-MAPK4 axis in suppressing NSCLC progression but also suggests a promising combination strategy for treating NSCLC.
Collapse
Affiliation(s)
- Shiqi Chen
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qian Hao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yu Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jing Tong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chen Xiong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Quan Liao
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Jiangxi, 330029, China
| | - Yang Zhang
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ting Ye
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Institute of Thoracic Oncology, Fudan University, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Xiang Zhou
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Haiquan Chen
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Institute of Thoracic Oncology, Fudan University, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
69
|
Liu Y, Stockwell BR, Jiang X, Gu W. p53-regulated non-apoptotic cell death pathways and their relevance in cancer and other diseases. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00842-3. [PMID: 40204927 DOI: 10.1038/s41580-025-00842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Programmed cell death is a mechanism that is crucial for numerous physiological and pathological processes. Whereas p53-mediated apoptosis is a major cell death pathway in cancer, accumulating evidence indicates that p53 also has crucial roles in controlling different non-apoptotic cell death (NACD) pathways, including ferroptosis, necroptosis, pyroptosis, autophagy-dependent cell death, entotic cell death, parthanatos and paraptosis, and may regulate PANoptosis, cuproptosis and disulfidptosis. Notably, the function of p53 in these NACDs substantially contributes to its biological effects, particularly in cancer development and other pathological processes. In this Review, we discuss recent advances in understanding the roles and underlying mechanisms of p53-mediated NACDs, focusing on ferroptosis, necroptosis and pyroptosis. We discuss the complex and distinct physiological settings in which NACDs are regulated by p53, and potential targeting of p53-regulated NACDs for the treatment of cancer and other human diseases. Finally, we highlight several important questions concerning p53-regulated NACDs that warrant further investigation.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, USA
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
70
|
Ren N, Chen H, Huang Y, Jin J, Zhang S, Yan R, Li M, Zheng L, Zou S, Li Y, Tan W, Lin D. MDM1 overexpression promotes p53 expression and cell apoptosis to enhance therapeutic sensitivity to chemoradiotherapy in patients with colorectal cancer. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0540. [PMID: 40200809 PMCID: PMC11976705 DOI: 10.20892/j.issn.2095-3941.2024.0540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/21/2025] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVE Identifying biomarkers that predict the efficacy and prognosis of chemoradiotherapy is important for individualized clinical treatment. We previously reported that high murine double minute 1 (MDM1) expression in patients with rectal cancer is linked to a favorable chemoradiation response. In this study the role of MDM1 in the chemoradiotherapy response in colorectal cancer (CRC) patients was evaluated. METHODS Colony formation and cell proliferation assays as well as xenograft models were used to determine if MDM1 expression affects the sensitivity of CRC cells to chemoradiation. RNA sequencing revealed that MDM1 regulates tumor protein 53 (TP53) expression and apoptosis. A series of molecular biology experiments were performed to determine how MDM1 affects p53 expression. The effects of inhibitors targeting apoptosis on MDM1 knockout cells were evaluated. RESULTS Gene expression profiling revealed that MDM1 is a potential chemoradiotherapy sensitivity marker. The sensitivity of CRC cells to chemoradiation treatment decreased after MDM1 knockout and increased after MDM1 overexpression. MDM1 affected p53 expression, thereby regulating apoptosis. MDM1 overexpression limited YBX1 binding to TP53 promoter, regulated TP53 expression, and rendered CRC cells more sensitive to chemoradiation. In CRC cells with low MDM1 expression, a combination of apoptosis-inducing inhibitors and chemoradiation treatment restored sensitivity to cancer therapy. CONCLUSIONS The current study showed that MDM1 expression influences the sensitivity of CRC cells to chemoradiation by influencing p53 and apoptosis pathways, which is the basis for the underlying molecular mechanism, and serves as a possible predictive marker for chemoradiotherapy prognosis.
Collapse
Affiliation(s)
- Ningxin Ren
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hongxia Chen
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ying Huang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518100, China
| | - Shaosen Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ruoqing Yan
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Mengjie Li
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Linlin Zheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuangmei Zou
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yexiong Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wen Tan
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dongxin Lin
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China
| |
Collapse
|
71
|
Ru Z, Li S, Wang M, Ni Y, Qiao H. Exploring Immune-Related Ferroptosis Genes in Thyroid Cancer: A Comprehensive Analysis. Biomedicines 2025; 13:903. [PMID: 40299520 PMCID: PMC12024864 DOI: 10.3390/biomedicines13040903] [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: 03/15/2025] [Accepted: 04/05/2025] [Indexed: 04/30/2025] Open
Abstract
Background: The increasing incidence and poor outcomes of recurrent thyroid cancer highlight the need for innovative therapies. Ferroptosis, a regulated cell death process linked to the tumour microenvironment (TME), offers a promising antitumour strategy. This study explored immune-related ferroptosis genes (IRFGs) in thyroid cancer to uncover novel therapeutic targets. Methods: CIBERSORTx and WGCNA were applied to data from TCGA-THCA to identify hub genes. A prognostic model composed of IRFGs was constructed using LASSO Cox regression. Pearson correlation was employed to analyse the relationships between IRFGs and immune features. Single-cell RNA sequencing (scRNA-seq) revealed gene expression in cell subsets, and qRT-PCR was used for validation. Results: Twelve IRFGs were identified through WGCNA, leading to the classification of thyroid cancer samples into three distinct subtypes. There were significant differences in patient outcomes among these subtypes. A prognostic risk score model was developed based on six key IRFGs (ACSL5, HSD17B11, CCL5, NCF2, PSME1, and ACTB), which were found to be closely associated with immune cell infiltration and immune responses within the TME. The prognostic risk score was identified as a risk factor for thyroid cancer outcomes (HR = 14.737, 95% CI = 1.95-111.65; p = 0.009). ScRNA-seq revealed the predominant expression of these genes in myeloid cells, with differential expression validated using qRT-PCR in thyroid tumour and normal tissues. Conclusions: This study integrates bulk and single-cell RNA sequencing data to identify IRFGs and construct a robust prognostic model, offering new therapeutic targets and improving prognostic evaluation for thyroid cancer patients.
Collapse
Affiliation(s)
- Zixuan Ru
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
| | - Siwei Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China;
| | - Minnan Wang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
| | - Yanan Ni
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
| | - Hong Qiao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
72
|
Zhang J, Chang K, Shangguan Y, Luo R, Bi Y, Yu Z, Li B. Flotillin- 1 ameliorates experimental diabetic retinopathy by inhibiting ferroptosis in blood-retinal barrier. J Mol Med (Berl) 2025:10.1007/s00109-025-02544-x. [PMID: 40198383 DOI: 10.1007/s00109-025-02544-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: 01/30/2025] [Revised: 03/07/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
Diabetic retinopathy (DR) is a chronic disease that severely impairs the vision of working individuals and is closely linked to blood-retinal barrier (BRB) dysfunction. Flotillin- 1 (FLOT1), a protein located in membrane lipid rafts, is essential for various intracellular biological processes. However, its role in the pathogenesis of DR remains unclear. Ferroptosis in high-glucose was assessed using Cell counting kit- 8 (CCK- 8), Malondialdehyde (MDA), Glutathione (GSH), Fe2+ assays, and transferrin expression. BRB disruption was evaluated with Evans blue staining. The interaction between FLOT1 and NF-E2-related factor 2 (Nrf2) was confirmed by immunoprecipitation and ferroptosis mechanisms were explored by inhibiting Nrf2 with ML385. In db/db mice (a type 2 diabetes model) was intravitreal injection of an adeno-associated virus (AAV) overexpressing FLOT1. Expression levels of Nrf2, solute carrier family 7 member 11 (SLC7 A11), and glutathione peroxidase 4 (GPX4) were evaluated in retina. Our study indicated that FLOT1 significantly alleviated BRB damage in DR, reversing high-glucose induced reductions in GPX4 and GSH, and inhibited the elevation of MDA and Fe2+. FLOT1 also suppressed ROS accumulation. Mechanistically, FLOT1 activates the Nrf2 pathway by enhancing its expression and promoting its nuclear translocation, thereby stimulating the SLC7 A11/GPX4 pathway to inhibiting lipid peroxidation and ferroptosis. We have identified ferroptosis is a key mechanism driving BRB damage in DR.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Ophthalmology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Ke Chang
- Department of Pharmacy, School of Medicine, Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Yanyu Shangguan
- Department of Ophthalmology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Ruoning Luo
- Department of Ophthalmology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Yanlong Bi
- Department of Ophthalmology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Zicheng Yu
- Department of Pharmacy, School of Medicine, Yangpu Hospital, Tongji University, Shanghai, 200090, China.
| | - Bing Li
- Department of Ophthalmology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
73
|
Ruiz CF, Ge X, McDonnell R, Agabiti SS, McQuaid DC, Tang A, Kharwa M, Goodell J, Saavedra-Peña RDM, Wing A, Li G, Medici NP, Robert ME, Varshney RR, Rudolph MC, Gorelick FS, Wysolmerski J, Canals D, Haley JD, Rodeheffer MS, Muzumdar MD. Diet-induced phospholipid remodeling dictates ferroptosis sensitivity and tumorigenesis in the pancreas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.645324. [PMID: 40235976 PMCID: PMC11996499 DOI: 10.1101/2025.04.04.645324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
High-fat diet (HFD) intake has been linked to an increased risk of pancreatic ductal adenocarcinoma (PDAC), a lethal and therapy-resistant cancer. However, whether and how specific dietary fats drive cancer development remains unresolved. Leveraging an oncogenic Kras -driven mouse model that closely mimics human PDAC progression, we screened a dozen isocaloric HFDs differing solely in fat source and representing the diversity of human fat consumption. Unexpectedly, diets rich in oleic acid - a monounsaturated fatty acid (MUFA) typically associated with good health - markedly enhanced tumorigenesis. Conversely, diets high in polyunsaturated fatty acids (PUFAs) suppressed tumor progression. Relative dietary fatty acid saturation levels (PUFA/MUFA) governed pancreatic membrane phospholipid composition, lipid peroxidation, and ferroptosis sensitivity in mice, concordant with circulating PUFA/MUFA levels being linked to altered PDAC risk in humans. These findings directly implicate dietary unsaturated fatty acids in controlling ferroptosis susceptibility and tumorigenesis, supporting potential "precision nutrition" strategies for PDAC prevention.
Collapse
|
74
|
Gupta H, Gupta A. Post-translational modifications of epigenetic modifier TIP60: their role in cellular functions and cancer. Epigenetics Chromatin 2025; 18:18. [PMID: 40186325 PMCID: PMC11969907 DOI: 10.1186/s13072-025-00572-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/27/2025] [Indexed: 04/07/2025] Open
Abstract
TIP60 is a crucial lysine acetyltransferase protein that catalyzes the acetylation of histone and non-histone proteins. This enzyme plays a crucial role in maintaining genomic integrity, by participating in DNA damage repair, ensuring accurate chromosomal segregation, and regulating a myriad of cellular processes such as apoptosis, autophagy, and wound-induced cell migration. One of the primary mechanisms through which TIP60 executes these diverse cellular functions is via post-translational modifications (PTMs). Over the years, extensive studies have demonstrated the importance of PTMs in controlling protein functions. This review aims to summarize the findings on PTMs occurring on the TIP60 protein and their functional implications. We also discuss previously uncharacterized PTM sites identified on TIP60 and examine their relationship with cancer-associated mutations, with a particular focus on residues potentially modified by various PTMs, to understand the cause of deregulation of TIP60 in various cancers.
Collapse
Affiliation(s)
- Himanshu Gupta
- Epigenetics and Human Disease Laboratory, Centre of Excellence in Epigenetics, Department of Life Sciences, Shiv Nadar Institution of Eminence, deemed to be University, Delhi-NCR, 201314, Uttar Pradesh, India
| | - Ashish Gupta
- Epigenetics and Human Disease Laboratory, Centre of Excellence in Epigenetics, Department of Life Sciences, Shiv Nadar Institution of Eminence, deemed to be University, Delhi-NCR, 201314, Uttar Pradesh, India.
| |
Collapse
|
75
|
Zhang N, Fan Y, Chen J, Gu J, Yan X. MAPK14 drives Ferroptosis and immune dysfunction in pediatric Sepsis-induced acute lung injury. Cell Immunol 2025; 411-412:104948. [PMID: 40209319 DOI: 10.1016/j.cellimm.2025.104948] [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: 02/15/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
OBJECTIVE Sepsis-induced acute lung injury (ALI) is driven by inflammation, oxidative stress, and immune suppression. MAPK14 (p38α) plays a role in ferroptosis and immune regulation, but its specific function in pediatric sepsis remains unclear. Therefore, our study aimed to explore the role and underlying mechanism of MAPK14 in pediatric sepsis. METHODS Bioinformatics analysis of GSE26440 and FerrDb identified ferroptosis-related genes in pediatric sepsis. STRING database was used to predict the proteins associated with MAPK14. MAPK14 expression in whole blood samples, LPS-treated MLE-12 cells, and a CLP mouse model was detected by qRT-PCR and western blot. Ferroptosis was assessed by measuring MDA, GSH, and Fe2+ levels, while ROS accumulation was analyzed using DCFH-DA staining and DHE staining. A cycloheximide (CHX) assay was performed to assess TTP53 protein stability. MPO immunohistochemistry and PD-L1 immunofluorescence assessed neutrophil infiltration, and flow cytometry evaluated neutrophil apoptosis. RESULTS Bioinformatics analysis of GSE26440 and FerrDb identified MAPK14 as a ferroptosis-related gene in pediatric sepsis. MAPK14 expression was upregulated in sepsis patient samples, LPS-treated MLE-12 cells and CLP mouse lung tissues. Overexpression of MAPK14 led to increased MDA and Fe2+ levels, reduced GSH, and elevated ROS fluorescence intensity, confirming its role in promoting ferroptosis. Mechanistically, MAPK14 upregulated TTP53, which in turn suppressed SLC7A11 and GPX4, further driving ferroptosis. MAPK14 overexpression stabilized TTP53 and enhanced its activity. Additionally, MAPK14 enhanced MPO and PD-L1 expression to promote neutrophil infiltration and immune suppression. Additionally, MAPK14 overexpression inhibited neutrophil apoptosis, promoted neutrophil infiltration and enhanced immune suppression. CONCLUSION MAPK14 drives ferroptosis via the TTP53/SLC7A11/GPX4 pathway and exacerbates immune suppression by promoting neutrophil infiltration.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Surgery Intensive Care Unit (SICU), Children's Hospital of Soochow University, Suzhou, Jiangsu 215008, China
| | - Yuanyuan Fan
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, China
| | - Juan Chen
- Nanjing Kangze Medical Testing Co., Ltd., Nanjing, Jiangsu 211100, China
| | - Juan Gu
- Department of Emergency, Yinchuan Maternal and Child Health Hospital, Yinchuan, Ningxia 750000, China
| | - Xiangming Yan
- Department of Urology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215008, China.
| |
Collapse
|
76
|
Haddadin L, Sun X. Stem Cells in Cancer: From Mechanisms to Therapeutic Strategies. Cells 2025; 14:538. [PMID: 40214491 PMCID: PMC11988674 DOI: 10.3390/cells14070538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Stem cells have emerged as a pivotal area of research in the field of oncology, offering new insights into the mechanisms of cancer initiation, progression, and resistance to therapy. This review provides a comprehensive overview of the role of stem cells in cancer, focusing on cancer stem cells (CSCs), their characteristics, and their implications for cancer therapy. We discuss the origin and identification of CSCs, their role in tumorigenesis, metastasis, and drug resistance, and the potential therapeutic strategies targeting CSCs. Additionally, we explore the use of normal stem cells in cancer therapy, focusing on their role in tissue regeneration and their use as delivery vehicles for anticancer agents. Finally, we highlight the challenges and future directions in stem cell research in cancer.
Collapse
Affiliation(s)
| | - Xueqin Sun
- Cancer Genome and Epigenetics Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
77
|
Li R, Wu X, Xue K, Wu S, Jiang G, He M, Xia Y, Liu H, Zhong M, Li J, Fan L, Li J. CircTADA2A stabilizes p53 via interacting with TRIM28 and suppresses the maintenance of FLT3-ITD acute myeloid leukemia. Leukemia 2025:10.1038/s41375-025-02589-4. [PMID: 40175625 DOI: 10.1038/s41375-025-02589-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Abstract
Internal tandem duplication mutations in the FMS-like tyrosine kinase 3 (FLT3-ITDs) occur in 25%-30% of acute myeloid leukemia (AML) cases and are associated with adverse prognosis. RNA-based therapeutics exhibit significant potential for treating diseases, prompting us to develop a novel circular RNA (circRNA)-based therapeutic strategy for FLT3-ITD AML. Here, we find circTADA2A is downregulated in FLT3-ITD AML patients. We further demonstrate that the downregulation of circTADA2A is critical for the proliferation of human FLT3-ITD AML cells, the sustenance of AML, and the self-renewal of leukemia stem/initiating cells (LSCs/LICs). Mechanistically, circTADA2A inhibits the TRIM28/MDM2 complexes formation by competitively binding to TRIM28, resulting in decreased levels of p53 ubiquitination and activating the p53 pathway. Importantly, in vitro transcription of circTADA2A and in vivo delivery via lipid nanoparticles (LNPs) significantly enhance the elimination of FLT3-ITD leukemia cells in combination with quizartinib treatment. In conclusion, our work uncovers the crucial functions of circTADA2A in the maintenance of FLT3-ITD AML and highlights a translationally important circTADA2A-based therapeutic approach for FLT3-ITD AML treatment.
Collapse
Affiliation(s)
- Ran Li
- Department of Hematology, Jiangsu Province Hospital, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolu Wu
- Department of Child Health Care, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Kai Xue
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shishuang Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ge Jiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengke He
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Xia
- Department of Hematology, Jiangsu Province Hospital, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Hailing Liu
- Department of Hematology, Jiangsu Province Hospital, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Miao Zhong
- Department of Hematology, Jiangsu Province Hospital, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jianyong Li
- Department of Hematology, Jiangsu Province Hospital, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Lei Fan
- Department of Hematology, Jiangsu Province Hospital, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Junmin Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
78
|
Cieślińska MW, Bialuk I, Dziemidowicz M, Szynaka B, Reszeć-Giełażyn J, Winnicka MM, Bonda TA. The Influence of Interleukin 6 Knockout on Age-Related Degenerative Changes in the Cerebellar Cortex of Mice. Cells 2025; 14:532. [PMID: 40214486 PMCID: PMC11989083 DOI: 10.3390/cells14070532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
This study investigates age-related neurodegeneration in the cerebellar cortex, emphasizing the role of IL-6 deficiency in preserving Purkinje cells. We found that apoptosis plays a minimal role in Purkinje cell loss by using 4-month- and 24-month-old wild-type (WT) and IL-6 knockout (IL-6KO) mice. At 24 months, WT mice exhibited severe Purkinje cell degeneration, including atrophic cell bodies, eosinophilic cytoplasm, pyknotic nuclei, mitochondrial disruption, and increased levels of lipofuscin-rich lysosomes. In contrast, IL-6KO mice showed fewer lysosomes, reduced mitochondrial damage, and less neuronal atrophy, indicating a neuroprotective effect. Lower p53 expression and decreased levels of its downstream effectors (p21, and Bax) in IL-6KO mice correlated with reduced cellular stress. Minimal changes in apoptotic markers (Bax and caspase-3) further reinforce the limited role of apoptosis. Neuroinflammation, marked by elevated GFAP, was prominent in aged WT mice but attenuated in IL-6KO mice. Reduced p53 accumulation, less severe neuroinflammation, and preserved metabolic homeostasis in IL-6KO mice correlated with improved Purkinje cell survival. These findings suggest that IL-6 accelerates neurodegeneration via p53-associated stress and inflammation, while IL-6 deficiency mitigates these effects. Targeting IL-6 signaling through anti-inflammatory strategies or IL-6 inhibition may offer a therapeutic approach for age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Magdalena Wiktoria Cieślińska
- Department of General and Experimental Pathology, Medical University of Białystok, Mickiewicza 2c, 15-222 Białystok, Poland
| | - Izabela Bialuk
- Department of General and Experimental Pathology, Medical University of Białystok, Mickiewicza 2c, 15-222 Białystok, Poland
| | - Magdalena Dziemidowicz
- Department of General and Experimental Pathology, Medical University of Białystok, Mickiewicza 2c, 15-222 Białystok, Poland
| | - Beata Szynaka
- Department of Histology and Embryology, Medical University of Bialystok, Waszyngtona 13, 15-269 Białystok, Poland
| | - Joanna Reszeć-Giełażyn
- Department of Medical Pathomorphology, Medical University of Bialystok, Waszyngtona 13, 15-269 Białystok, Poland
| | - Maria Małgorzata Winnicka
- Department of General and Experimental Pathology, Medical University of Białystok, Mickiewicza 2c, 15-222 Białystok, Poland
| | - Tomasz Andrzej Bonda
- Department of General and Experimental Pathology, Medical University of Białystok, Mickiewicza 2c, 15-222 Białystok, Poland
| |
Collapse
|
79
|
Zheng B, Liu K, Feng J, Ouyang Q, Jia T, Wang Y, Tian S, Chen X, Cai T, Wen L, Zhang X, Li X, Ma X. GAMT facilitates tumor progression via inhibiting p53 in clear cell renal cell carcinoma. Biol Direct 2025; 20:43. [PMID: 40176130 PMCID: PMC11966922 DOI: 10.1186/s13062-025-00641-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/23/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the most common type of RCC. Even though the targeted drugs for the treatment of ccRCC have a certain therapeutic effect, due to the problem of drug resistance, the search for new targets for targeted therapy of ccRCC remains urgent. GAMT is an enzyme involved in creatine metabolism. However, the precise biological roles and molecular mechanisms of GAMT in ccRCC are not fully understood. RESULTS Here, we found that GAMT was upregulated in ccRCC cells and tissues and associated with poor prognosis. Further, GAMT has pro-oncogenic abilities in promoting ccRCC development and progression. Intriguingly, GAMT exerted biological functions independent of its role in catalyzing creatine synthesis. Mechanistically, GAMT overexpression contributes to the development and progression of ccRCC by inhibiting tumor suppressor p53. Finally, we identified fisetin as a novel GAMT inhibitor and validated its role in suppressing ccRCC progression and sensitizing ccRCC cells to targeted drug axitinib via in vivo and in vitro assays. CONCLUSIONS This study reveals that GAMT has pro-oncogenic abilities in promoting ccRCC development and progression. GAMT exerted its non-enzymatic functions possibly by regulating the expression of p53. Fisetin, the novel GAMT inhibitor identified herein, may serve as a new antitumor drug for ccRCC treatment.
Collapse
Affiliation(s)
- Bin Zheng
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Kan Liu
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Ji Feng
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Qing Ouyang
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Tongyu Jia
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yaohui Wang
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Shuo Tian
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Xinran Chen
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Tianwei Cai
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Lequan Wen
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Xu Zhang
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiubin Li
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xin Ma
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
80
|
Zuo X, Wang X, Xie J, Jia Y. Emodin alleviates the damage to lens epithelial cells in diabetic cataract by repressing the p53-mediated ferroptosis pathway. Int Ophthalmol 2025; 45:141. [PMID: 40175804 DOI: 10.1007/s10792-025-03513-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/12/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Diabetic cataract (DC) is an ocular complication caused by diabetes. Currently, the main treatments for DC include pharmacological therapy and surgical intervention. The core objective of this study is to elucidate the specific mechanism of action of emodin in the treatment of DC, thereby providing potential targets for the treatment of DC. METHODS CCK-8 kit was used to detect the effect of emodin on the activity of lens epithelial cells (LECs). The impact of emodin on the expression of inflammatory factors and apoptosis in high glucose-induced LECs were evaluated by utilizing ELISA and flow cytometry. Then, commercial kits were performed to detect the regulatory effects of emodin on oxidative stress and ferroptosis in high glucose LECs. The potential mechanism of emodin in combating DC by inhibiting ferroptosis was analyzed by network pharmacology methods, and protein binding activity to emodin was measured by molecular docking. Besides, western blot (WB) assay was used to detect the effect of emodin on p53. RESULTS Firstly, the results of CCK-8 showed that emodin could effectively alleviate the decrease of LECs cell activity and Lactate dehydrogenase (LDH) release induced by high glucose. Emodin suppressed high glucose-induced apoptosis of LECs, reduced the release of inflammatory factors, and alleviated oxidative stress and ferroptosis. GO and KEGG analyses confirmed the involvement of oxidative stress (OS), inflammatory response, and ferroptosis in the process of emodin treatment for DC. Molecular docking studies showed that emodin stably bound to proteins such as TP53, TNF, IL-6, and IL-1β. Additionally, WB results indicated that emodin alleviated high glucose-induced ferroptosis by binding to p53. CONCLUSION Collectively, these data suggest that emodin alleviates damage to LECs by interfering with the p53-mediated ferroptosis pathway, thereby attenuating DC disease, which offered new directions for the development of new drugs.
Collapse
Affiliation(s)
- Xiangrong Zuo
- Department of Ophthalmology, Xingtai People's Hospital, No.818 Xiangdu North Road, Xingtai City, 054000, Hebei, China.
| | - Xiuxian Wang
- Department of Ophthalmology, Xingtai People's Hospital, No.818 Xiangdu North Road, Xingtai City, 054000, Hebei, China
| | - Jing Xie
- Department of Ophthalmology, Xingtai People's Hospital, No.818 Xiangdu North Road, Xingtai City, 054000, Hebei, China
| | - Yuhong Jia
- Department of Ophthalmology, Xingtai People's Hospital, No.818 Xiangdu North Road, Xingtai City, 054000, Hebei, China
| |
Collapse
|
81
|
Zhu Y, Yu Y, Jia Y, Lin Z, Lei J, Wu D, Xu T, Li L, Zheng B. The Characteristics and Functions of Orally Absorbed Herbal Decoction-Borne Plant MicroRNAs. PLANTA MEDICA 2025; 91:283-292. [PMID: 39875131 DOI: 10.1055/a-2527-2127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Herbal decoctions always contain numerous plant microRNAs, and some of these can be absorbed orally to exert cross-kingdom gene regulation. However, little is known about which specific types of herbal decoction-borne plant microRNAs are more likely to be absorbed. Thus, two antiviral herbal decoctions, Qingfei Paidu and Qingre Huashi Kangdu, were administered to human volunteers and rats, respectively, to investigate the characteristics of orally absorbed decoction-borne plant microRNAs. MIR-6240 - 3 p was identified as an absorbed plant microRNA in humans and is most highly expressed in Qingfei Paidu decoction. Therefore, the kinetics of MIR-6240 - 3 p were monitored in humans following the administration of the Qingfei Paidu decoction, and its antiviral effect on human coronavirus type 229E (HCoV-229E) was examined in vitro. There were 586 176 small RNAs identified in Qingfei Paidu decoction, of which 100 276 were orally absorbed by humans. In the Qingre Huashi Kangdu decoction, 124 026 small RNAs were detected, with 7484 being orally absorbed by rats. Logistical repression analysis revealed that absorbable plant small RNAs in both humans and rats presented higher expression levels, greater minimum free energy, and increased AU/UA frequencies compared to nonabsorbable plant small RNAs. The amount of MIR-6240 - 3 p in humans increased between 1 and 3 h after the administration of the Qingfei Paidu decoction. In addition, MIR-6240 - 3 p significantly reduced the RNA copy number and TCID50 of HCoV-229E in vitro. These results suggest that herbal decoction-borne plant small RNAs with a higher expression level, greater minimum free energy, or an increased AU/UA frequency are more likely to be orally absorbed and could potentially mediate cross-kingdom gene regulation.
Collapse
Affiliation(s)
- Yating Zhu
- Evidence-Based Medicine Research Center, Jiangxi University of Chinese Medicine, Nanchang City, Jiangxi Province, P. R. China
| | - Yicheng Yu
- Affiliated Hospital, Jiangxi University of Chinese Medicine, Nanchang City, Jiangxi Province, P. R. China
| | - Yao Jia
- Evidence-Based Medicine Research Center, Jiangxi University of Chinese Medicine, Nanchang City, Jiangxi Province, P. R. China
| | - Ziqi Lin
- Evidence-Based Medicine Research Center, Jiangxi University of Chinese Medicine, Nanchang City, Jiangxi Province, P. R. China
| | - Jinyue Lei
- Evidence-Based Medicine Research Center, Jiangxi University of Chinese Medicine, Nanchang City, Jiangxi Province, P. R. China
| | - Diyao Wu
- Evidence-Based Medicine Research Center, Jiangxi University of Chinese Medicine, Nanchang City, Jiangxi Province, P. R. China
| | - Tielong Xu
- Evidence-Based Medicine Research Center, Jiangxi University of Chinese Medicine, Nanchang City, Jiangxi Province, P. R. China
| | - Longxue Li
- Laboratory Animal Science and Technology Center, Jiangxi University of Chinese Medicine, Nanchang City, Jiangxi Province, P. R. China
| | - Bin Zheng
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, and National Center for Tropical Diseases Research, Shanghai, P. R. China
| |
Collapse
|
82
|
Mete M, Ojha A, Dhar P, Das D. Deciphering Ferroptosis: From Molecular Pathways to Machine Learning-Guided Therapeutic Innovation. Mol Biotechnol 2025; 67:1290-1309. [PMID: 38613722 DOI: 10.1007/s12033-024-01139-0] [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: 12/10/2023] [Accepted: 03/11/2024] [Indexed: 04/15/2024]
Abstract
Ferroptosis is a unique form of cell death reliant on iron and lipid peroxidation. It disrupts redox balance, causing cell death by damaging the plasma membrane, with inducers acting through enzymatic pathways or transport systems. In cancer treatment, suppressing ferroptosis or circumventing it holds significant promise. Beyond cancer, ferroptosis affects aging, organs, metabolism, and nervous system. Understanding ferroptosis mechanisms holds promise for uncovering novel therapeutic strategies across a spectrum of diseases. However, detection and regulation of this regulated cell death are still mired with challenges. The dearth of cell, tissue, or organ-specific biomarkers muted the pharmacological use of ferroptosis. This review covers recent studies on ferroptosis, detailing its properties, key genes, metabolic pathways, and regulatory networks, emphasizing the interaction between cellular signaling and ferroptotic cell death. It also summarizes recent findings on ferroptosis inducers, inhibitors, and regulators, highlighting their potential therapeutic applications across diseases. The review addresses challenges in utilizing ferroptosis therapeutically and explores the use of machine learning to uncover complex patterns in ferroptosis-related data, aiding in the discovery of biomarkers, predictive models, and therapeutic targets. Finally, it discusses emerging research areas and the importance of continued investigation to harness the full therapeutic potential of targeting ferroptosis.
Collapse
Affiliation(s)
- Megha Mete
- Department of Bioengineering, National Institute of Technology Agartala, Agartala, Tripura, 799046, India
| | - Amiya Ojha
- Department of Bioengineering, National Institute of Technology Agartala, Agartala, Tripura, 799046, India
| | - Priyanka Dhar
- CSIR-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Deeplina Das
- Department of Bioengineering, National Institute of Technology Agartala, Agartala, Tripura, 799046, India.
| |
Collapse
|
83
|
Han Z, Wang Y, Zang X, Liu H, Su J, Zhou Y. FePt/MnO 2@PEG Nanoparticles as Multifunctional Radiosensitizers for Enhancing Ferroptosis and Alleviating Hypoxia in Osteosarcoma Therapy. IEEE Trans Nanobioscience 2025; 24:180-190. [PMID: 39392735 DOI: 10.1109/tnb.2024.3475051] [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: 10/13/2024]
Abstract
Radiotherapy (RT) is a widely used cancer treatment, and the use of metal-based nanoradiotherapy sensitizers has demonstrated promise in enhancing its efficacy. However, achieving effective accumulation of these sensitizers within tumors and overcoming resistance induced by the hypoxic tumor microenvironment remain challenging issues. In this study, we developed FePt/MnO2@PEG nanoparticles with multiple radiosensitizing mechanisms, including high-atomic-number element-mediated radiation capture, catalase-mimicking oxygenation, and GSH depletion-induced ferroptosis. Both in vitro and in vivo experiments were conducted to validate the radiosensitizing mechanisms and therapeutic efficacy of FePt/MnO2@PEG. In conclusion, this study presents a novel and clinically relevant strategy and establishes a safe and effective combination radiotherapy approach for cancer treatment. These findings hold significant potential for improving radiotherapy outcomes and advancing the field of nanomedicine in cancer therapy.
Collapse
|
84
|
Yin X, Liu Z, Li C, Wang J. Hinokitiol ameliorates MASH in mice by therapeutic targeting of hepatic Nrf2 and inhibiting hepatocyte ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156472. [PMID: 39922149 DOI: 10.1016/j.phymed.2025.156472] [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: 08/06/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH), an advanced stage of metabolic dysfunction-associated steatotic liver disease (MASLD), still lacks approved effective clinical drugs. Ferroptosis, a form of regulated cell death driven by excessive iron accumulation and uncontrollable lipid peroxidation, has been proven to be a trigger of inflammation and initiation of steatohepatitis. The pathogenic interplay is modulated by oxidative stress, while the Nrf2-mediated antioxidant response plays a regulatory role in ferroptosis. Phytochemical hinokitiol (Hino) has demonstrated positive efficacy in hepatocellular carcinoma (HCC) in the reported work, but it remains unknown whether its therapeutic effect attributes to delaying the progress of steatohepatitis to HCC. PURPOSE This work aimed to systemically investigate the significance of ferroptosis in the pathogenesis of MASH and to demonstrate that Hino, a bioactive monoterpene compound, attenuates the primary pathological characteristics of MASH via promotion of Nrf2/GPX4 signaling. METHODS In this work, a MASH model was established using the high-fat/high-cholesterol (HFHC) diet-fed in vivo and palmitic acid/oleic acid (PO)-stimulated hepatocytes in vitro. Biochemical indexes, pathological analysis, western blot, PCR assay, energy metabolic phenotype, molecular docking, and confirmatory assays were performed comprehensively to reveal the key link between the Nrf2/GPX4 axis and the treatment of MASH. RESULTS Under MASH conditions with increased oxidative stress, we show that Nrf2 was remarkable downregulated in HFHC diet-fed mice and PO-managed hepatocytes. Mechanistically, hepatic upregulation of Nrf2 through phytochemical Hino supplementation inhibited ferroptosis, enhanced lipid metabolism, and thereby alleviated hepatic steatosis, inflammation, and fibrosis. Conversely, silencing Nrf2 in hepatocytes further promoted the accumulation of key markers of ferroptosis and aggravated MASH phenotypes. CONCLUSION Increased ferroptosis promoted steatosis which further drove inflammation and hepatic fibrosis. Our results suggested the significance of Nrf2 in ameliorating MASH, which was regulated through Hino. Thus, targeted inhibition of ferroptosis through Hino administration is a feasible and effective approach for treating MASH.
Collapse
Affiliation(s)
- Xunzhe Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, No.5625, Ren Min Street, Changchun, Jilin 130022, China; Center for Theoretical Interdisciplinary Sciences, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Zuojia Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, No.5625, Ren Min Street, Changchun, Jilin 130022, China.
| | - Chang Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, No.5625, Ren Min Street, Changchun, Jilin 130022, China
| | - Jin Wang
- Center for Theoretical Interdisciplinary Sciences, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China; Department of Chemistry and Physics, Stony Brook University, Stony Brook, New York 11794-3400, USA.
| |
Collapse
|
85
|
Peng B, Zhang J, Xiang Y. CDCA8 and its multifaceted role in tumorigenesis. Biomed Pharmacother 2025; 185:117951. [PMID: 40056827 DOI: 10.1016/j.biopha.2025.117951] [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: 02/11/2025] [Accepted: 02/27/2025] [Indexed: 03/10/2025] Open
Abstract
Human cell division cycle-associated 8 (CDCA8), also known as Borealin or Dasra-B, is a critical component of the vertebrate Chromosomal Passenger Complex (CPC). It plays a pivotal role in the segregation of sister chromatids during the cell cycle and is essential for preventing the formation of aneuploid chromosomes and ensuring successful cytokinesis. Numerous studies have demonstrated that CDCA8 is upregulated in various cancers, including hepatocellular carcinoma, lung cancer, glioma, and bladder cancer. By influencing key biological processes such as cell proliferation, apoptosis, invasion, and metastasis, CDCA8 drives tumor progression. Clinically, the expression of CDCA8 correlates closely with tumor staging and histological grading, providing significant prognostic value for patients with diverse cancers. Moreover, CDCA8 modulates tumor biology through multiple signaling pathways, including P53, PI3K/Akt, E2F/Rb, and mTOR. In summary, CDCA8 represents a promising diagnostic and therapeutic target across multiple cancer types and serves as a potential prognostic biomarker. This review highlights the critical roles of CDCA8 in cancer diagnosis, treatment, and prognosis, as well as the underlying mechanisms through which it exerts its effects. These insights offer a theoretical basis and research direction for early cancer diagnosis, targeted therapy, and prognostic evaluation.
Collapse
Affiliation(s)
- Boming Peng
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| | - Jianquan Zhang
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China; Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou 570208, China.
| | - Yang Xiang
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China; Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou 570208, China
| |
Collapse
|
86
|
Crawford CEW, Burslem GM. Acetylation: a new target for protein degradation in cancer. Trends Cancer 2025; 11:403-420. [PMID: 40055119 PMCID: PMC11981854 DOI: 10.1016/j.trecan.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 04/11/2025]
Abstract
Acetylation is an increasing area of focus for cancer research as it is closely related to a variety of cellular processes through modulation of histone and non-histone proteins. However, broadly targeting acetylation threatens to yield nonselective toxic effects owing to the vital role of acetylation in cellular function. There is thus a pressing need to elucidate and characterize the specific cancer-relevant roles of acetylation for future therapeutic design. Acetylation-mediated protein homeostasis is an example of selective acetylation that affects a myriad of proteins as well as their correlated functions. We review recent examples of acetylation-mediated protein homeostasis that have emerged as key contributors to tumorigenesis, tumor proliferation, metastasis, and/or drug resistance, and we discuss their implications for future exploration of this intriguing phenomenon.
Collapse
Affiliation(s)
- Callie E W Crawford
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - George M Burslem
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA; Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA.
| |
Collapse
|
87
|
Cui X, Chen B, Chen Y, Zhou M, Cao L, Hu B, Wu J, Ma X, Ying T. Engineering Iridium Nanoclusters for Boosting Ferroptotic Cell Death by Regulating GPX4 and p53 Functions. Adv Healthc Mater 2025; 14:e2404895. [PMID: 40018815 DOI: 10.1002/adhm.202404895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/17/2025] [Indexed: 03/01/2025]
Abstract
Emerging evidence indicates that modulating glutathione peroxidase 4 (GPX4) to induce ferroptosis is a promising strategy for tumor treatment. However, most of the GPX4 small molecule inhibitors face limitations due to their poor delivery efficacy and low specificity of ferroptosis activation. Herein, a ferroptosis-inducing nanomedicine is developed that integrates nutlin-3 with iridium oxide nanoclusters (NUT-IrOx NCs) for enhanced ferroptosis-driven multimodal therapeutic efficacy in colorectal cancer (CRC). This NUT-IrOx NCs can induce glutathione (GSH) depletion via enhanced Ir (VI)-Ir (III) transition, while nutlin-3, a well-established inhibitor of the p53-MDM2 interaction, suppresses GSH production by modulation of the p53/SLC7A11/xCT signaling pathway. The reduction of intracellular GSH results in pronounced reductions of GPX4 enzymatic activity, consequently leading to lipid peroxidation accumulation and further enhancing ferroptosis-induced CRC therapy. This dual-pronged approach demonstrates robust anticancer therapeutic effects with favorable biocompatibility in both in vitro and in vivo CRC models. This study provides an effective strategy that highlights the benefits of inhibiting of GSH/GPX4 by activating multiple ferroptosis regulatory pathways, providing an alternative therapeutic avenue for CRC treatment.
Collapse
Affiliation(s)
- Xiaoyu Cui
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, P. R. China
| | - Bin Chen
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong, University School of Medicine, Shanghai, 200233, P. R. China
| | - Ying Chen
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong, University School of Medicine, Shanghai, 200233, P. R. China
| | - Mi Zhou
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong, University School of Medicine, Shanghai, 200233, P. R. China
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Lijun Cao
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, P. R. China
| | - Bing Hu
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong, University School of Medicine, Shanghai, 200233, P. R. China
| | - Jianrong Wu
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong, University School of Medicine, Shanghai, 200233, P. R. China
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Xinxin Ma
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong, University School of Medicine, Shanghai, 200233, P. R. China
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Tao Ying
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, P. R. China
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong, University School of Medicine, Shanghai, 200233, P. R. China
| |
Collapse
|
88
|
Koo KY, Moon K, Song HS, Lee MS. Metabolic regulation by p53: Implications for cancer therapy. Mol Cells 2025; 48:100198. [PMID: 39986611 PMCID: PMC11925517 DOI: 10.1016/j.mocell.2025.100198] [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: 12/16/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025] Open
Abstract
The tumor suppressor p53, long known for its roles in maintaining genomic integrity and suppressing tumorigenesis, has recently been recognized as a key regulator of cellular metabolism. Here, we review p53's emerging metabolic functions, highlighting its ability to orchestrate glucose, amino acid, and lipid metabolism. By promoting oxidative phosphorylation while inhibiting glycolysis and anabolic pathways, wild-type p53 counters metabolic reprogramming characteristic of cancer cells, such as the Warburg effect, and protects cells from mild cellular stresses. In contrast, mutant p53 disrupts these processes, fostering metabolic adaptations that support tumor progression. These findings pave the way for therapeutic approaches targeting p53-driven metabolic vulnerabilities in cancer.
Collapse
Affiliation(s)
- Ki Yeon Koo
- Department of Life Sciences, POSTECH, Pohang, Korea
| | - Kwanho Moon
- Department of Life Sciences, POSTECH, Pohang, Korea
| | | | - Min-Sik Lee
- Department of Life Sciences, POSTECH, Pohang, Korea.
| |
Collapse
|
89
|
Altea-Manzano P, Decker-Farrell A, Janowitz T, Erez A. Metabolic interplays between the tumour and the host shape the tumour macroenvironment. Nat Rev Cancer 2025; 25:274-292. [PMID: 39833533 DOI: 10.1038/s41568-024-00786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Metabolic reprogramming of cancer cells and the tumour microenvironment are pivotal characteristics of cancers, and studying these processes offer insights and avenues for cancer diagnostics and therapeutics. Recent advancements have underscored the impact of host systemic features, termed macroenvironment, on facilitating cancer progression. During tumorigenesis, these inherent features of the host, such as germline genetics, immune profile and the metabolic status, influence how the body responds to cancer. In parallel, as cancer grows, it induces systemic effects beyond the primary tumour site and affects the macroenvironment, for example, through inflammation, the metabolic end-stage syndrome of cachexia, and metabolic dysregulation. Therefore, understanding the intricate metabolic interplay between the tumour and the host is a growing frontier in advancing cancer diagnosis and therapy. In this Review, we explore the specific contribution of the metabolic fitness of the host to cancer initiation, progression and response to therapy. We then delineate the complex metabolic crosstalk between the tumour, the microenvironment and the host, which promotes disease progression to metastasis and cachexia. The metabolic relationships among the host, cancer pathogenesis and the consequent responsive systemic manifestations during cancer progression provide new perspectives for mechanistic cancer therapy and improved management of patients with cancer.
Collapse
Affiliation(s)
| | | | | | - Ayelet Erez
- Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
90
|
Liu C, Liu G, Zhou F, Chen L, Chang B, Tang H, Wang H. EBF1-induced CSRP2 boosts the progression of B-cell acute lymphocytic leukemia by inhibiting ferroptosis. Cancer Lett 2025; 614:217556. [PMID: 39952599 DOI: 10.1016/j.canlet.2025.217556] [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: 01/09/2025] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
B-cell acute lymphocytic leukemia (B-ALL) is a highly aggressive malignancy with poor prognosis. Developing diagnostic markers and therapeutic targets to identify and treat B-ALL early would improve the outcomes of B-ALL patients. Here, we conducted RNA next-generation sequencing using bone marrow (BM) specimens obtained from 7 B-ALL patients and 7 healthy donors. We found cysteine and glycine-rich protein 2 (CSRP2) upregulated in B-ALL. Down-regulation of CSRP2 resulted in suppressed cell proliferation and enhanced cell apoptosis in B-ALL. In addition, inhibition of CSRP2 increased cell ferroptosis in B-ALL cells. Mechanically, we revealed that transcription factor early B cell factor 1 (EBF1) regulated CSRP2 levels in B-ALL, and inhibition of EBF1 decreased CSRP2 levels in B-ALL. In conclusion, the dysregulation EBF1 led to CSRP2 upregulation and resulting in progression of B-ALL. The EBF1/CSRP2 axis could be of great potential as therapeutic targets for B-ALL treatment.
Collapse
Affiliation(s)
- Chengcheng Liu
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen Institute of Hematology, Guangzhou, Guangdong, 510630, PR China
| | - Gexiu Liu
- Institute of Hematology, Jinan University, Guangzhou, Guangdong, 510632, PR China
| | - Fenling Zhou
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen Institute of Hematology, Guangzhou, Guangdong, 510630, PR China; Institute of Hematology, Jinan University, Guangzhou, Guangdong, 510632, PR China
| | - Lu Chen
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen Institute of Hematology, Guangzhou, Guangdong, 510630, PR China; Institute of Hematology, Jinan University, Guangzhou, Guangdong, 510632, PR China
| | - Boyang Chang
- Department of Interventional Radiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, PR China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| | - Hua Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| |
Collapse
|
91
|
Zheng J, Conrad M. Ferroptosis: when metabolism meets cell death. Physiol Rev 2025; 105:651-706. [PMID: 39661331 DOI: 10.1152/physrev.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
We present here a comprehensive update on recent advancements in the field of ferroptosis, with a particular emphasis on its metabolic underpinnings and physiological impacts. After briefly introducing landmark studies that have helped to shape the concept of ferroptosis as a distinct form of cell death, we critically evaluate the key metabolic determinants involved in its regulation. These include the metabolism of essential trace elements such as selenium and iron; amino acids such as cyst(e)ine, methionine, glutamine/glutamate, and tryptophan; and carbohydrates, covering glycolysis, the citric acid cycle, the electron transport chain, and the pentose phosphate pathway. We also delve into the mevalonate pathway and subsequent cholesterol biosynthesis, including intermediate metabolites like dimethylallyl pyrophosphate, squalene, coenzyme Q (CoQ), vitamin K, and 7-dehydrocholesterol, as well as fatty acid and phospholipid metabolism, including the biosynthesis and remodeling of ester and ether phospholipids and lipid peroxidation. Next, we highlight major ferroptosis surveillance systems, specifically the cyst(e)ine/glutathione/glutathione peroxidase 4 axis, the NAD(P)H/ferroptosis suppressor protein 1/CoQ/vitamin K system, and the guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin/dihydrofolate reductase axis. We also discuss other potential anti- and proferroptotic systems, including glutathione S-transferase P1, peroxiredoxin 6, dihydroorotate dehydrogenase, glycerol-3-phosphate dehydrogenase 2, vitamin K epoxide reductase complex subunit 1 like 1, nitric oxide, and acyl-CoA synthetase long-chain family member 4. Finally, we explore ferroptosis's physiological roles in aging, tumor suppression, and infection control, its pathological implications in tissue ischemia-reperfusion injury and neurodegeneration, and its potential therapeutic applications in cancer treatment. Existing drugs and compounds that may regulate ferroptosis in vivo are enumerated.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, Garching, Germany
| |
Collapse
|
92
|
Yang Y, Nguyen D, Chen K, Zeng FG. Evaluating synthesized speech intelligibility in noise. JASA EXPRESS LETTERS 2025; 5:045202. [PMID: 40243623 DOI: 10.1121/10.0036397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025]
Abstract
Humans can modify their speech to improve intelligibility in noisy environments. With the advancement of speech synthesis technology, machines may also synthesize voices that remain highly intelligible in noise condition. This study evaluates both the subjective and objective intelligibility of synthesized speech in speech-shaped noise from three major speech synthesis platforms. It was found that synthesized voices have a similar intelligibility range to human voices, and some synthesized voices were more intelligible than human voices. It was also found that two modern automatic speech recognition systems recognized 10% more words than human listeners.
Collapse
Affiliation(s)
- Ye Yang
- Department of Biomedical Engineering, University of California, Irvine, California 92697, USA
| | - Dathan Nguyen
- Department of Biomedical Engineering, University of California, Irvine, California 92697, USA
| | - Katherine Chen
- Department of Biomedical Engineering, University of California, Irvine, California 92697, USA
| | - Fan-Gang Zeng
- Department of Biomedical Engineering, University of California, Irvine, California 92697, USA
- Department of Otolaryngology - Head and Neck Surgery, University of California, Irvine, California 92697, , , ,
| |
Collapse
|
93
|
Chen X, Rong Y, Jiang Y, Zhang Q, Xiang S, Chen Z, Chen W, Zhang H, Deng C, Wang J. Vitamin K1 Alleviates Retinal Inflammation Following Acute Ocular Hypertension by Modulating Microglial Ferroptosis. Invest Ophthalmol Vis Sci 2025; 66:46. [PMID: 40244608 PMCID: PMC12013678 DOI: 10.1167/iovs.66.4.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/13/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose Glaucoma is the leading cause of irreversible blindness worldwide and encompasses a group of diseases characterized by optic nerve atrophy and visual field defects. Acute intraocular pressure (IOP) elevation is a key driver of retinal inflammation and optic nerve damage, often accompanied by microglial activation and dysregulated ferroptosis pathways. Vitamin K1, a fat-soluble vitamin, possesses anti-inflammatory and antioxidant properties, and has the potential to regulate ferroptosis. However, its mechanisms in alleviating retinal inflammation following acute IOP elevation remain unclear. Methods In vivo, we established a mouse model of acute ocular hypertension to evaluate the protective effects of vitamin K1 on the retina and visual function. Transcriptome sequencing was used to explore the underlying mechanisms by which vitamin K1 exerts its effects. Immunofluorescence and Western blot were used to assess retinal inflammation and observe ferroptosis in microglia. In vitro, we developed a BV2 cell OGDR model to investigate the regulatory effects of vitamin K1 on iron metabolism and inflammation in microglia. Results Our findings demonstrated that acute IOP elevation led to microglial activation, along with iron overload and ferroptosis in microglia. Further analyses revealed that microglial ferroptosis was accompanied by an upregulation of inflammatory cytokine gene expression and protein levels. Vitamin K1 intervention, however, inhibited microglial ferroptosis, alleviated retinal inflammation, minimized retinal ganglion cell (RGC) loss, and protected visual function. Conclusions In conclusion, this study demonstrates that vitamin K1 exerts a protective effect by modulating microglial ferroptosis, thereby alleviating acute ocular hypertension-induced retinal inflammation.
Collapse
Affiliation(s)
- Xi Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Rong
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuxian Jiang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiuxiang Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sifei Xiang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiqi Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chaohua Deng
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junming Wang
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
94
|
Yao M, Zhu X, Chen YC, Yang GH, Ao P. Exploring Multi-Target Therapeutic Strategies for Glioblastoma via Endogenous Network Modeling. Int J Mol Sci 2025; 26:3283. [PMID: 40244148 PMCID: PMC11989339 DOI: 10.3390/ijms26073283] [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: 01/14/2025] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Medical treatment of glioblastoma presents a significant challenge. A conventional medication has limited effectiveness, and a single-target therapy is usually effective only in the early stage of the treatment. Recently, there has been increasing focus on multi-target therapies, but the vast range of possible combinations makes clinical experimentation and implementation difficult. From the perspective of systems biology, this study conducted simulations for multi-target glioblastoma therapy based on dynamic analysis of previously established endogenous networks, validated with glioblastoma single-cell RNA sequencing data. Several potentially effective target combinations were identified. The findings also highlight the necessity of multi-target rather than single-target intervention strategies in cancer treatment, as well as the promise in clinical applications and personalized therapies.
Collapse
Affiliation(s)
- Mengchao Yao
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai 200444, China;
| | - Xiaomei Zhu
- Shanghai Key Laboratory of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200444, China
| | - Yong-Cong Chen
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai 200444, China;
| | - Guo-Hong Yang
- Department of Physics, Shanghai University, Shanghai 200444, China;
| | - Ping Ao
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
95
|
Yuan F, Han S, Li Y, Li S, Li D, Tian Q, Feng R, Shao Y, Liang X, Wang J, Lei H, Li X, Duan Y. miR-214-3p attenuates ferroptosis-induced cellular damage in a mouse model of diabetic retinopathy through the p53/SLC7A11/GPX4 axis. Exp Eye Res 2025; 253:110299. [PMID: 39978746 DOI: 10.1016/j.exer.2025.110299] [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: 12/17/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Ferroptosis has been implicated in the development of diabetic retinopathy (DR). This study aimed to identify novel ferroptosis-related regulators involved in the pathophysiology of DR using an in vivo streptozotocin (STZ)-induced diabetic model in C57BL/6J mice and cultured primary human retinal vascular endothelial cells (HRECs). Transmission electron microscopy revealed mitochondrial morphological changes consistent with ferroptosis in vascular endothelial cells from STZ-treated mice. Western blot analysis showed increased levels of ferroptosis markers (4-HNE, p53, phosphorylated p53) along with decreased levels of glutathione (GSH), SLC7A11, and GPX4 in diabetic mice. In vitro experiments demonstrated that ferroptosis inhibitors, including pifithrin-α (a p53 inhibitor) and ferrostatin-1 (Fer-1), mitigated cellular damage and Fe2+ accumulation in high-glucose-treated HRECs. These inhibitors also improved mitochondrial membrane potential and restored GSH levels. Bioinformatics analysis and dual-luciferase assays identified a p53 binding site within the miR-214-3p sequence. Overexpression of miR-214-3p in high-glucose-treated HRECs resulted in downregulation of p53 and upregulation of SLC7A11 and GPX4, thereby alleviating ferroptosis-induced injury. This study demonstrates that ferroptosis contributes to retinal damage at tissue, cellular, and molecular levels in DR. Specifically, p53, regulated by miR-214-3p, promotes ferroptosis through the SLC7A11/GPX4 pathway under high-glucose conditions. These findings suggest that the miR-214-3p/p53/SLC7A11/GPX4 axis could serve as a potential therapeutic target for managing ferroptosis and retinal damage in diabetic retinopathy.
Collapse
Affiliation(s)
- Fang Yuan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China; Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Songyu Han
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yahong Li
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sanming Li
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Dian Li
- Washington University in St. Louis, MO, USA
| | - Qingjun Tian
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ronghua Feng
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Shao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xing Liang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingbo Wang
- Shanxi Eye Hospital Affiliated with Shanxi Medical University, Taiyuan, 030072, China
| | - Hetian Lei
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.
| | - Yajian Duan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China; Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
96
|
Cui W, Hao M, Yang X, Yin C, Chu B. Gut microbial metabolism in ferroptosis and colorectal cancer. Trends Cell Biol 2025; 35:341-351. [PMID: 39261152 DOI: 10.1016/j.tcb.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/13/2024]
Abstract
Ferroptosis is programmed cell death induced by iron-driven lipid peroxidation. Numerous studies have shown that ferroptosis is implicated in the progression of colorectal cancer (CRC) and has emerged as a promising strategy to combat therapy-resistant CRC. While the intrinsic antiferroptotic and proferroptotic pathways in CRC cells have been well characterized, extrinsic metabolism pathways regulating ferroptosis in CRC pathogenesis remain less understood. Emerging evidence shows that gut microbial metabolism is tightly correlated with the progression of CRC. This review provides an overview of gut microbial metabolism and discusses how these metabolites derived from intestinal microflora contribute to cancer plasticity through ferroptosis. Targeting gut microbe-mediated ferroptosis is a potential approach for CRC treatment.
Collapse
Affiliation(s)
- Weiwei Cui
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Meng Hao
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Xin Yang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Chengqian Yin
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China.
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
97
|
Liu X, Wang W, Nie Q, Liu X, Sun L, Ma Q, Zhang J, Wei Y. The Role and Mechanisms of Ubiquitin-Proteasome System-Mediated Ferroptosis in Neurological Disorders. Neurosci Bull 2025; 41:691-706. [PMID: 39775589 PMCID: PMC11979074 DOI: 10.1007/s12264-024-01343-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/29/2024] [Indexed: 01/11/2025] Open
Abstract
Ferroptosis is a form of cell death elicited by an imbalance in intracellular iron concentrations, leading to enhanced lipid peroxidation. In neurological disorders, both oxidative stress and mitochondrial damage can contribute to ferroptosis, resulting in nerve cell dysfunction and death. The ubiquitin-proteasome system (UPS) refers to a cellular pathway in which specific proteins are tagged with ubiquitin for recognition and degradation by the proteasome. In neurological conditions, the UPS plays a significant role in regulating ferroptosis. In this review, we outline how the UPS regulates iron metabolism, ferroptosis, and their interplay in neurological diseases. In addition, we discuss the future application of small-molecule inhibitors and identify potential drug targets. Further investigation into the mechanisms of UPS-mediated ferroptosis will provide novel insights and strategies for therapeutic interventions and clinical applications in neurological diseases.
Collapse
Affiliation(s)
- Xin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Wei Wang
- Cancer Biology Institute, Baotou Medical College, Baotou, 014010, China
| | - Qiucheng Nie
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xinjing Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Lili Sun
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qiang Ma
- Cancer Biology Institute, Baotou Medical College, Baotou, 014010, China
| | - Jie Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Yiju Wei
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
98
|
Huan X, Li J, Chu Z, Zhang H, Cheng L, Lun P, Du X, Chen X, Jiao Q, Jiang H. Dysregulation of Iron Homeostasis Mediated by FTH Increases Ferroptosis Sensitivity in TP53-Mutant Glioblastoma. Neurosci Bull 2025; 41:569-582. [PMID: 39666195 PMCID: PMC11978602 DOI: 10.1007/s12264-024-01322-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/19/2024] [Indexed: 12/13/2024] Open
Abstract
Iron metabolism is a critical factor in tumorigenesis and development. Although TP53 mutations are prevalent in glioblastoma (GBM), the mechanisms by which TP53 regulates iron metabolism remain elusive. We reveal an imbalance iron homeostasis in GBM via TCGA database analysis. TP53 mutations disrupted iron homeostasis in GBM, characterized by elevated total iron levels and reduced ferritin (FTH). The gain-of-function effect triggered by TP53 mutations upregulates itchy E3 ubiquitin-protein ligase (ITCH) protein expression in astrocytes, leading to FTH degradation and an increase in free iron levels. TP53-mut astrocytes were more tolerant to the high iron environment induced by exogenous ferric ammonium citrate (FAC), but the increase in intracellular free iron made them more sensitive to Erastin-induced ferroptosis. Interestingly, we found that Erastin combined with FAC treatment significantly increased ferroptosis. These findings provide new insights for drug development and therapeutic modalities for GBM patients with TP53 mutations from iron metabolism perspectives.
Collapse
Affiliation(s)
- Xuejie Huan
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Jiangang Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Zhaobin Chu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Hongliang Zhang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Lei Cheng
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Peng Lun
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
- Qingdao Key Laboratory of Neurorehabilitation, University of Health and Rehabilitation Sciences, Qingdao, 266113, China.
| |
Collapse
|
99
|
Song W, Yuan Y, Cao F, Pan H, Liu Y. The key pathways and genes related to oncolytic Newcastle disease virus-induced phenotypic changes in ovarian cancer cells. J Microbiol 2025; 63:e2411018. [PMID: 40313149 DOI: 10.71150/jm.2411018] [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: 11/12/2024] [Accepted: 01/24/2025] [Indexed: 05/03/2025]
Abstract
The poor prognosis and high recurrence rate of ovarian cancer highlight the urgent need to develop new therapeutic strategies. Oncolytic Newcastle disease virus (NDV) can kill cancer cells directly and regulate innate and adaptive immunity. In this study, ovarian cancer cells infected with or without velogenic NDV-BJ were subjected to a CCK-8 assay for detecting cell proliferation, flow cytometry for detecting the cell cycle and apoptosis, and wound healing and transwell assays for detecting cell migration and invasion. Transcriptomic sequencing was conducted to identify the differentially expressed genes (DEGs). GO and KEGG enrichment analyses were performed to explore the mechanism underlying the oncolytic effect of NDV on ovarian cancer cells. The results showed that infection with NDV inhibited ovarian cancer cell proliferation, migration, and invasion; disrupted the cell cycle; and promoted apoptosis. Compared with those in negative control cells, the numbers of upregulated and downregulated genes in ovarian cancer cells infected with NDV were 1,499 and 2,260, respectively. Thirteen KEGG pathways related to cell growth and death, cell mobility, and signal transduction were significantly enriched. Among these pathways, 48 DEGs, especially SESN2, HLA B/C/E, GADD45B, and RELA, that may be involved in the oncolytic process were screened, and qPCR analysis verified the reliability of the transcription data. This study discovered some key pathways and genes related to oncolytic NDV-induced phenotypic changes in ovarian cancer cells, which will guide our future research directions and help further explore the specific mechanisms by which infection with NDV suppresses ovarian cancer development.
Collapse
Affiliation(s)
- Wei Song
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao 266000, P. R. China
| | - Yuan Yuan
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao 266000, P. R. China
| | - Fangfang Cao
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao 266000, P. R. China
| | - Huazheng Pan
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao 266000, P. R. China
| | - Yaqing Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao 266000, P. R. China
| |
Collapse
|
100
|
Mao N, Zhang M, Shen M, Yuan J, Lin Z. Research progress on ferroptosis in cerebral hemorrhage. Biomed Pharmacother 2025; 185:117932. [PMID: 40015051 DOI: 10.1016/j.biopha.2025.117932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/06/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
The pathophysiology of intracerebral hemorrhage (ICH) is complex and can cause variable degrees of cell death. Recently, ferroptosis, an emerging cell death mechanism, has garnered significant attention in cerebral hemorrhage disorder. This study aimed to examine iron mortality after cerebral hemorrhage and current targets for potential therapeutic interventions. We specifically focused on iron metabolism abnormalities, lipid peroxidation, and related neuroinflammation and introduced molecular mechanisms, including transcription factors, to gain a better understanding of the underlying mechanisms of ferroptosis and investigate possible therapeutic options for ICH.
Collapse
Affiliation(s)
- Niping Mao
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Zhang
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming Shen
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junhui Yuan
- Department of Neonatology, Wenling maternal and child health care hospital, Wenling, Zhejiang, China.
| | - Zhenlang Lin
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|