1
|
Miao M, Chen Y, Wang X, Li S, Hu R. The critical role of ferroptosis in virus-associated hematologic malignancies and its potential value in antiviral-antitumor therapy. Virulence 2025; 16:2497908. [PMID: 40302035 PMCID: PMC12045570 DOI: 10.1080/21505594.2025.2497908] [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: 12/03/2024] [Revised: 03/06/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
Epstein-Barr Virus (EBV), Kaposi's sarcoma-associated herpesvirus (KSHV), and human T-cell leukemia virus type 1 (HTLV-1) are key infectious agents linked to the development of various hematological malignancies, including Hodgkin's lymphoma, non-Hodgkin's lymphoma, and adult T-cell leukemia/lymphoma. This review highlights the critical knowledge gaps in understanding the role of ferroptosis, a novel form of cell death, in virus-related tumors. We focus on how ferroptosis influences the host cell response to these viral infections, revealing groundbreaking mechanisms by which the three viruses differentially regulate core pathways of ferroptosis, such as iron homeostasis, lipid peroxidation, and antioxidant systems, thereby promoting malignant transformation of host cells. Additionally, we explore the potential of antiviral drugs and ferroptosis modulators in the treatment of virus-associated hematological malignancies.
Collapse
Affiliation(s)
- Miao Miao
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuelei Chen
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Xuehan Wang
- Shenyang Shenhua Institute Test Technology, Shenyang, Liaoning, China
| | - Shengyang Li
- Publishing Department, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Rong Hu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Guo R, Wang R, Zhang W, Li Y, Wang Y, Wang H, Li X, Song J. Multifaceted regulatory mechanisms of the EGR family in tumours and prospects for therapeutic applications (Review). Int J Mol Med 2025; 56:113. [PMID: 40444475 PMCID: PMC12121985 DOI: 10.3892/ijmm.2025.5554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 05/14/2025] [Indexed: 06/02/2025] Open
Abstract
The early growth response (EGR) family comprises four zinc finger transcription factors: EGR1, EGR2, EGR3 and EGR4. These transcription factors belong to the Cys2‑His2‑type zinc finger protein family and are essential in cell differentiation, proliferation, apoptosis and stress response. Initially, EGR1 was recognised for its essential regulatory role in tumourigenesis. Recent studies have identified similarities between other members of the EGR family and EGR1 in tumour regulation and the multifaceted regulatory mechanism employed by the EGR family to affect tumours. Therefore, the present review describes the dual roles of the EGR family in tumours and their regulatory mechanisms in immunity, metabolism and differentiation. Additionally, the present review offers a new perspective on relevant tumour therapeutic studies based on current EGR targeting.
Collapse
Affiliation(s)
- Rongqi Guo
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Rui Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Weisong Zhang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Yangyang Li
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Yihao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Hao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Xia Li
- Department of General Medicine, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Jianxiang Song
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| |
Collapse
|
3
|
Deng Y, Zhong L, Zhao Y, Wan P, Zhang Y, Liao Y, Zhang H, Wang M, Liu B. A Dendrobium chrysotoxum extract erianin induce AML cells death by activating PPARɑ and downregulating PI3K/AKT signaling pathways. Toxicon 2025; 261:108371. [PMID: 40280444 DOI: 10.1016/j.toxicon.2025.108371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Erianin is a biphenyl compound with low toxicity and a single structure that is extracted from Dendrobium officinale. The wide spectrum of pharmacological properties and excellent toxicity of erianin have been comprehensively proven in multiple tumors. However, less is known about the toxicity of erianin in acute myeloid leukemia (acute myeloid leukemia AML). Here, we explored the anti-AML capacity and potential mechanisms of erianin. Cells proliferation and cytotoxicity of AML cells of erianin was detected by CCK-8 assay and flow cytometer was conducted to assess AML cells apoptosis rate. Erianin blocked the AML cells cycle at the G2/M phase by regulating cell cycle-related protein and P21, P27, and P53 mRNA expression. Additionally, we first filtered PPARɑ and PIK3R1 through network pharmacology, protein-protein interaction (PPI) network, and GO and KEGG pathway enrichment analysis and confirmed their binding with erianin by molecular docking analysis.The cellular thermal shift assay (CETSA) and the drug affinity responsive target stability assay (DARTS) further verified that PPARɑ was an effective target of erianin. Specifically, erianin was found to inhibit the transcriptional level of PIK3R1 by promoting the protein expression of PPARɑ, thereby inhibiting the PI3K/AKT pathway. The inhibitory effect of erianin was partially neutralized by GW6471, a PPARɑ inhibitor. Notably, erianin revealed vigoroso coordinate repression with LY294002 on AML cells. Our findings indicate that erianin showed a potent cytotoxic effect on AML cells and affected AML cells via PPARɑ to regulate PI3K/AKT signaling pathways. We demonstrated the potent anti-AML effects of erianin and reported its potential mechanisms of action, indicating its potential for further development as a novel anti-AML drug.
Collapse
Affiliation(s)
- Ying Deng
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Liang Zhong
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, China
| | - Yi Zhao
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Peng Wan
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Ying Zhang
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Yang Liao
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, China
| | - Hongyan Zhang
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Meng Wang
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Beizhong Liu
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, China; Clinical Laboratory of the Affiliated Rehabilitation Hospital, Chongqing Medical University, China.
| |
Collapse
|
4
|
Li S, Liu Z, Chen Y, Feng S, Chen H, Zhao Y, He Y, Wang Q. Repaglinide platinum(IV) conjugates: Enhancing p53 signaling for antitumor and antimetastatic efficacy. J Inorg Biochem 2025; 268:112910. [PMID: 40199143 DOI: 10.1016/j.jinorgbio.2025.112910] [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: 01/04/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
The tumor suppressor p53 plays multiple roles at the crossroads of suppressing tumor development and metastasis. Here, a series of Repaglinide platinum(IV) conjugates promoting the p53 pathway were designed and prepared, which displayed potent antiproliferative and antimetastatic activities both in vitro and in vivo. Mechanistically, the expression of p53 was upregulated by the synergistic functions of the platinum core through causing severe DNA damage, and the RPG ligand via stimulating the lumican/p53/p21 pathway. The mitochondria-mediated apoptosis was initiated, involving the Bcl-2/Bax/caspase pathway. Pro-death autophagy was initiated with the upregulation of LC3II and down regulation of p62. Additionally, angiogenesis was suppressed by reversing tumor inflammation through the inhibition of key enzymes COX-2, MMP9, and VEGFA. Furthermore, antitumor immunity was enhanced by blocking the immune checkpoint PD-L1, which led to an increased presence of CD3+ and CD8+ T-cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Suying Li
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Yan Chen
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Shuaiqi Feng
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Hengye Chen
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Yanna Zhao
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Yanqin He
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China.
| |
Collapse
|
5
|
Nakada T, Koga M, Takeuchi H, Doi K, Sugiyama H, Sakurai H. PP2A adapter protein IER5 induces dephosphorylation and degradation of MDM2, thereby stabilizing p53. Cell Signal 2025; 131:111739. [PMID: 40081547 DOI: 10.1016/j.cellsig.2025.111739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/11/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
The tumor suppressor p53 activates transcription of the IER5 gene, which encodes an adapter protein of protein phosphatase PP2A. IER5 binds to both the B55 regulatory subunit of PP2A and PP2A's target proteins, facilitating PP2A/B55-catalyzed dephosphorylation of these proteins. Here, we show that IER5 functions as a positive regulator of p53 by inhibiting its ubiquitination, thereby increasing cellular p53 levels. Mechanistically, this effect of IER5 requires its nuclear localization and binding to both PP2A/B55 and the p53 ubiquitin E3 ligase MDM2. Importantly, IER5 fails to inhibit p53 ubiquitination in cells treated with the MDM2 inhibitor Nutlin-3. The IER5-PP2A/B55 complex dephosphorylates MDM2 at Ser166, leading to MDM2 ubiquitination and a reduction in nuclear MDM2. Altogether, our data provide evidence that IER5-PP2A/B55 regulates the nuclear balance between MDM2 and p53 via MDM2 dephosphorylation.
Collapse
Affiliation(s)
- Taisei Nakada
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Mayuko Koga
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Hiroto Takeuchi
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Kuriko Doi
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Haruka Sugiyama
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Hiroshi Sakurai
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan.
| |
Collapse
|
6
|
Chen XL, Ojalill M, Jean C, Tancioni I, Jiang S, Boyer A, Ozmadenci D, Uryu S, Tarin D, Schlessinger J, Stupack DG, Schlaepfer DD. Inducible FAK loss but not FAK inhibition in endothelial cells of PYK2-null mice activates p53 tumor suppressor to prevent tumor growth. Mol Biol Cell 2025; 36:ar64. [PMID: 40202821 DOI: 10.1091/mbc.e24-12-0562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
Focal adhesion kinase (FAK) and the related tyrosine kinase PYK2 are signaling and scaffolding proteins co-expressed in endothelial cells (ECs) that regulate blood vessel function and tumor growth. As FAK-PYK2 share overlapping cellular roles, we generated PYK2-/- FAKfl/fl mice with tamoxifen-inducible EC-specific Cre expression. EC FAK inactivation in PYK2-/- but not PYK2+/+ mice led to increased heart and lung mass, vascular leakage, and created a tumor microenvironment that was repressive to syngeneic melanoma, breast, and lung carcinoma implanted tumor growth. Tumor suppression was associated with defective vessel sprouting, enhanced p53 tumor suppressor and p21CIP1 protein expression in ECs, elevated markers of DNA damage, and altered blood cytokine levels in tumor-bearing mice. However, EC-specific hemizygous kinase-defective (KD) FAK expression in EC FAK-/KD PYK2-/- mice was not associated with elevated p53 levels. Instead, EC FAK-/KD PYK2-/- mice supported primary tumor growth but prevented metastasis, implicating EC FAK activity in tumor spread. In vitro, combined genetic or small molecule FAK-PYK2 knockdown in ECs or tumor cells elevated p21CIP1 and prevented cell proliferation in a p53-dependent manner, highlighting a linkage between EC FAK-PYK2 loss and p53 activation in tumor regulation.
Collapse
Affiliation(s)
- Xiao Lei Chen
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Marjaana Ojalill
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Christine Jean
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Isabelle Tancioni
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Shulin Jiang
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Antonia Boyer
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Duygu Ozmadenci
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Sean Uryu
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - David Tarin
- Department of Pathology, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Joseph Schlessinger
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
| | - Dwayne G Stupack
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - David D Schlaepfer
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| |
Collapse
|
7
|
Luo Y, Yu Y, Zeng F, Yi Y, Lu Z, Lin B, Chen L, Zeng Z, Luo D, Liu A. Acetylation of FABP3 alleviates radioimmunotherapy-induced cardiomyocyte senescence by modulating long-chain polyunsaturated fatty acid metabolism. Int Immunopharmacol 2025; 160:114912. [PMID: 40449275 DOI: 10.1016/j.intimp.2025.114912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 05/06/2025] [Accepted: 05/17/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND The combination of thoracic radiotherapy and immunotherapy (radioimmunotherapy) has shown significant antitumor efficacy but is associated with increased cardiotoxicity, the mechanisms of which remain poorly understood. METHODS A total of 72 male C57BL/6 J mice were employed to establish the radioimmunotherapy-induced cardiac injury model, with 18 mice allocated to each of four groups, including the IR group (single-dose 16 Gy cardiac irradiation), ICI group (PD-1 inhibitor 200 μg every 3 days), iRT group (16 Gy cardiac irradiation combined with PD-1 inhibitor), and Control group (IgG). Cardiac function and myocardial senescence were assessed at 28 days, 3 months, and 5 months post-intervention. Additionally, myocardial tissue transcriptomics, non-targeted metabolomics, and acetylated proteomics were performed at 28 days post-intervention, integrated with molecular experiments to investigate the mechanisms of cardiomyocyte senescence. H9C2 cardiomyocytes with FABP3 K45 acetylation-mimetic (K45Q), empty vector (EV), and non-acetylatable (K45R) mutant were used for functional validation. RESULTS Combined radioimmunotherapy significantly exacerbated cardiac dysfunction and cardiomyocyte senescence in murine models, manifested with elevated serum levels of cardiac injury biomarkers of cTnI and NT-proBNP, reduced LVEF and LVFS, aggravated myocardial histopathological changes characterized by enhanced inflammatory infiltration, interstitial edema, and myocardium structure disorder in iRT group compared to the other three groups. Concomitantly, compared with other groups, the senescence-associated markers (p16, p21, and SASP factors) in the myocardial tissues of the iRT group were markedly upregulated from 28 days to 5 months. By integrating transcriptomic and non-targeted metabolomics analyses, as well as molecular experiments, we revealed that radioimmunotherapy resulted in dysregulated myocardial metabolism by suppressing ATP production, promoting lipid droplet accumulation, mitochondrial dysfunction, and fatty acid metabolism alterations, particularly involving long-chain polyunsaturated fatty acid (PUFAs) metabolism. Acetylome profiling identified a significant increase in FABP3 K45 acetylation (log2FC = 8.73, P < 0.05) in iRT vs. Control group, with acute-phase elevation (28 days, P < 0.001) and chronic-phase reduction (3 months, P < 0.001). Functional validation in H9C2 cardiomyocytes demonstrated that, compared to EV and K45R groups, FABP3 K45Q attenuated cellular senescence, enhanced mitochondrial oxidative phosphorylation, fatty acid metabolism, and ATP production, while attenuated ROS generation, lipid droplet accumulation, and glycolysis. Metabolomic analysis also revealed the acetylation of FABP3 K45 was significantly associated with the synthesis or accumulation of PUFAs, such as arachidonic acid and linoleic acid, which may alleviate cardiomyocyte senescence by enhancing energy supply and blocking the synthesis of inflammatory mediators. CONCLUSION FABP3 K45 acetylation mitigates radioimmunotherapy-induced cardiomyocyte senescence and metabolic dysfunction, revealing a novel regulatory mechanism that links post-translational modifications to cardiac cellular homeostasis under combined radioimmunotherapy.
Collapse
Affiliation(s)
- Yuxi Luo
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Ying Yu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Fujuan Zeng
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Yali Yi
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Zhiqin Lu
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Bilin Lin
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Leifeng Chen
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Zhimin Zeng
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province 330006, China.
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China.
| |
Collapse
|
8
|
Wang M, Han D, Xiang L, Wang J, Luo B, Shen X. 20-Hydroxyecdysone Modulates Bmp53-Mediated Apoptosis Regulation by Suppressing Mdm2-like-Dependent Ubiquitination in Silkworm, Bombyx mori. INSECTS 2025; 16:544. [PMID: 40429257 PMCID: PMC12112081 DOI: 10.3390/insects16050544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
In the silkworm, 20-hydroxyecdysone (20E) induces apoptosis and autophagy, driving larval organ degeneration and remodeling. This mechanism may be a potential target for eco-friendly strategy for insect pests. However, a major challenge in harnessing this approach lies in the insufficient understanding of 20E's regulatory process in such a cell death mechanism. Our previous research has identified Bmp53 as a crucial gene in promoting the development of Bombyx mori during the pupal stage by inducing apoptosis, and has predicted the potential apoptotic regulatory network of Bmp53, wherein Mdm2-like ubiquitinating structural protein serves as a key component of this network. This study demonstrates that Mdm2-like acts as a ubiquitination regulatory protein, controlling its apoptosis-inducing activity via interaction with Bmp53. Moreover, co-expression of Mdm2-like and Bmp53 indirectly affects gene expression in the 20E-mediated apoptosis pathway. Further investigation revealed that Mdm2-like suppresses 20E-induced apoptosis by downregulating Bmp53 expression. This study reveals that the ubiquitination-mediated Mdm2-like/Bmp53 apoptosis pathway is a novel mechanism regulating silkworm apoptosis, with 20E playing a crucial role in this process. These findings enhance our understanding of the genetic mechanisms underlying tissue degradation during the metamorphic stage of the Bombyx mori. Additionally, these insights provide a theoretical reference for the development of environmentally friendly, hormone-based control strategies targeting protein modification as a means of managing lepidopteran pests.
Collapse
Affiliation(s)
- Meixian Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (M.W.); (D.H.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Dingding Han
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (M.W.); (D.H.)
| | - Luyang Xiang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (M.W.); (D.H.)
| | - Jiahao Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (M.W.); (D.H.)
| | - Benteng Luo
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (M.W.); (D.H.)
| | - Xingjia Shen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (M.W.); (D.H.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| |
Collapse
|
9
|
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; 124:1658-1673. [PMID: 40221836 DOI: 10.1016/j.bpj.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
10
|
Fang W, Chen Y, Nie M, Zhou X, Liu Y, Tao H, Yang B, Wang X. Targeting YY1-DR5 Axis by Pyripyropene O as a Novel Therapeutic Strategy Against Prostate Cancer: Molecular Mechanisms and In Vivo Zebrafish Validation. Mar Drugs 2025; 23:214. [PMID: 40422804 DOI: 10.3390/md23050214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Induction of apoptosis is an important strategy for the treatment of prostate cancer. DR5 is a member of the death receptor superfamily and targeting DR5 is an effective way to induce apoptosis. Pyripyropene O is a natural compound isolated from the marine fungus Aspergillus fumigatus SCSIO 41220. We found it has anti-prostate cancer potential by inducing apoptosis; Methods: The effects of pyripyropene O on the viability, proliferation, cell cycle, apoptosis and migration of prostate cancer cells were investigated by MTT assay, plate clone formation assay, 3D cell sphere assay, flow cytometry and real-time cell analysis. Transmission electron microscopy was used to observe the changes in the internal structure of prostate cancer cells after treatment with pyripyropene O. After determining the mode of cell death, the mechanism of action of pyripyropene O on prostate cancer was further investigated using apoptotic protein microarray, western blot, qPCR, molecular docking, cellular immunofluorescence staining and cellular thermal shift assay. After explaining the mechanism of action of pyriproxyfen O, the in vivo absorption, distribution, metabolism, excretion and potential toxicity of pyriproxyfen O were investigated using ADMETLab 2.0 software. Finally, a zebrafish xenograft tumour model was developed to evaluate the anti-prostate cancer effects of pyriproxyfen O in vivo; Results: The experimental results at the cellular level showed that pyripyropene O inhibited the survival, proliferation and migration of prostate cancer cells, and also showed that pyripyropene O blocked the prostate cancer cell cycle at the G2/M phase and induced apoptosis. At the molecular level, pyripyropene O binds to the transcription factor YY1, promotes YY1 nuclear translocation, regulates the transcription level of DR5, a target gene of YY1, and upregulates the expression of DR5 mRNA and protein. The in vivo results showed that pyripyropene O effectively inhibited the development of prostate cancer in zebrafish; Conclusions: Pyripyropene O has a clear anti-prostate cancer effect at both cellular and animal levels, inhibiting the survival and proliferation of prostate cancer cells by binding to the transcription factor YY1 to activate the expression of DR5 to promote apoptosis, thus exerting an inhibitory effect on prostate cancer.
Collapse
Affiliation(s)
- Wenxuan Fang
- Guangxi Engineering Research Center for High-Value Utilization of Guangxi-Produced Authentic Medicinal Herbs, Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Ying Chen
- Guangdong Key Laboratory of Marine Materia Medica/State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Mingyi Nie
- Guangxi Engineering Research Center for High-Value Utilization of Guangxi-Produced Authentic Medicinal Herbs, Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Xuefeng Zhou
- Guangdong Key Laboratory of Marine Materia Medica/State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Yonghong Liu
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
- Guangdong Key Laboratory of Marine Materia Medica/State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Huaming Tao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Bin Yang
- Guangdong Key Laboratory of Marine Materia Medica/State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Xueni Wang
- Guangxi Engineering Research Center for High-Value Utilization of Guangxi-Produced Authentic Medicinal Herbs, Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| |
Collapse
|
11
|
Yan H, Huang J, Wang Y, Zhang Y, Ren W, Zhai Z, Tang Y, Lai H, Fan X, Liu L, Leung ELH. Berbamine as potential STING inhibitor For KRAS-mutant non-small cell lung cancer. Pharmacol Res 2025; 216:107777. [PMID: 40383171 DOI: 10.1016/j.phrs.2025.107777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/27/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
Lung adenocarcinoma (LUAD) is a leading cause of cancer-related mortality. Poor prognostic results in LUAD are frequently associated with KRAS mutations and drug resistance. KRAS mutations can induce STING activation by triggering DNA damage response (DDR) activation. This persistently activated STING signaling gives rise to an immunosuppressive microenvironment, thereby complicating treatment efforts. In this study, we identified that the low-toxicity pro-apoptotic drug Berbamine (BBM) as a potential therapeutic agent for LUAD cells with KRAS mutations. BBM exhibits anti-tumor effects by triggering cell cycle arrest, enhancing senescence, and activating apoptosis. BBM also targets STING, leading to the downregulation of p-STING (Ser366) and CCL2. This in turn reduced the infiltration of M-MDSCs into the tumor microenvironment. These combined mechanisms not only suppress STING-dependent tumor growth but also remodel the immunosuppressive tumor microenvironment, thereby enhancing anti-tumor immunity. Collectively, our findings position BBM as a promising therapeutic agent for LUAD with KRAS mutations, offering a strategy to target STING-associated pathways, overcome immune suppression, and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Haoxin Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao
| | - Jumin Huang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao
| | - Yuwei Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province 712046, China
| | - Yizhong Zhang
- Chinese Medicine and Translational Medicine R&D center, Zhuhai UM Science & Technology Research Institute, Zhuhai, Guangdong 519031, China
| | - Wenkang Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao
| | - Zhiran Zhai
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao
| | - Yuping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province 712046, China
| | - Huanling Lai
- Guangzhou National Laboratory, Guangzhou, Guangdong 510000, China
| | - Xingxing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao.
| | - Lihua Liu
- School of Economics and Management, Yanbian University, Yanji 133000, China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao.
| |
Collapse
|
12
|
Baldassarre G, L de la Serna I, Vallette FM. Death-ision: the link between cellular resilience and cancer resistance to treatments. Mol Cancer 2025; 24:144. [PMID: 40375296 PMCID: PMC12080166 DOI: 10.1186/s12943-025-02339-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025] Open
Abstract
One of the key challenges in defeating advanced tumors is the ability of cancer cells to evade the selective pressure imposed by chemotherapy, targeted therapies, immunotherapy and cellular therapies. Both genetic and epigenetic alterations contribute to the development of resistance, allowing cancer cells to survive initially effective treatments. In this narration, we explore how genetic and epigenetic regulatory mechanisms influence the state of tumor cells and their responsiveness to different therapeutic strategies. We further propose that an altered balance between cell growth and cell death is a fundamental driver of drug resistance. Cell death programs exist in various forms, shaped by cell type, triggering factors, and microenvironmental conditions. These processes are governed by temporal and spatial constraints and appear to be more heterogeneous than previously understood. To capture the intricate interplay between death-inducing signals and survival mechanisms, we introduce the concept of Death-ision. This framework highlights the dynamic nature of cell death regulation, determining whether specific cancer cell clones evade or succumb to therapy. Building on this understanding offers promising strategies to counteract resistant clones and enhance therapeutic efficacy. For instance, combining DNMT inhibitors with immune checkpoint blockade may counteract YAP1-driven resistance or the use of transcriptional CDK inhibitors could prevent or overcome chemotherapy resistance. Death-ision aims to provide a deeper understanding of the diversity and evolution of cell death programs, not only at diagnosis but also throughout disease progression and treatment adaptation.
Collapse
Affiliation(s)
- Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, 33081, Italy.
| | - Ivana L de la Serna
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| | - François M Vallette
- Centre de Recherche en Cancérologie et Immunologie Intégrées Nantes Angers (CRCI2 NA), INSERM UMR1307/CNRS UMR 6075/Nantes Université/Univ. Angers. Nantes, 44007, Nantes, France.
- Institut de Cancérologie de L'Ouest (ICO), 44085, Saint-Herblain, France.
| |
Collapse
|
13
|
Lindström MS. The Central Role of Ribosomal Proteins in p53 Regulation. Cancers (Basel) 2025; 17:1597. [PMID: 40427096 PMCID: PMC12110047 DOI: 10.3390/cancers17101597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
The tumor suppressor protein p53 prevents the malignant transformation of cells by responding to DNA damage, oncogene activation, and abnormal growth signals including ribosome assembly defects. Under normal conditions, p53 activity is controlled by the regulatory proteins MDM2 and MDM4, which suppress its function through ubiquitin-mediated degradation and transcriptional inhibition. A subset of ribosomal proteins initiates the p53 response to impaired ribosome biogenesis. The ability of some ribosomal proteins to control MDM2 and MDM4 activities, and thereby p53, underscores an intriguing aspect of cell biology: proteins primarily known for their roles in ribosome function can exert extra-ribosomal functions. One notable example is the cellular RNA-protein complex involving RPL5, RPL11, and 5S rRNA (5S RNP) which inhibits MDM2 and stabilizes p53. Another RP, RPL22, is frequently mutated in cancers with microsatellite instability and its paralog RPL22L1 is often amplified. Recent studies have revealed that RPL22 directly modulates the alternative splicing of MDM4 to promote p53 activation, suggesting that the ribosomal protein-p53 relationship is more complex than previously thought. Cellular responses to ribosome biogenesis inhibition extend beyond general alterations in transcription and translation to actively determine cancer cell fate by selectively engaging tumor-suppressor pathways. RPL22's effect on MDM4 and other mRNA splicing events is a striking example. A better understanding of the mechanisms involved could guide the development of improved cancer treatments.
Collapse
Affiliation(s)
- Mikael S Lindström
- Department of Medical Biochemistry and Biophysics, Division of Genome Biology, Science for Life Laboratory, Karolinska Institutet, SE-171 21 Stockholm, Sweden
| |
Collapse
|
14
|
Zhang Y, Su W, Ji X, Yang Z, Guan Q, Pang Y, Zhong L, Wang Y, Xiang J. PCSK9 promotes progression of anaplastic thyroid cancer through E-cadherin endocytosis. Cell Death Dis 2025; 16:362. [PMID: 40328788 PMCID: PMC12056021 DOI: 10.1038/s41419-025-07690-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 04/04/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
Although anaplastic thyroid cancer (ATC) constitutes only 1-2% of all thyroid malignancies, it is associated with an exceptionally high mortality rate, accounting for 14-39% of thyroid cancer-related deaths. In this study, we identified the critical role of Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) in ATC progression. Proteomic analysis revealed E-cadherin as a key mediator of PCSK9-driven malignancy in ATC. Mechanistically, PCSK9 promotes the degradation of E-cadherin through the lysosomal pathway. Furthermore, the loss of the p53 function, particularly the R248Q mutation, de-repressed PCSK9 expression at the transcriptional level. Notably, the PCSK9 inhibitor PF-846 considerably suppressed ATC proliferation and metastasis in both in vitro and in vivo models. In conclusion, PCSK9 enhances ATC malignancy by regulating E-cadherin degradation via the lysosomal pathway, underscoring its potential as a promising therapeutic target.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wei Su
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaoyu Ji
- Department of Oncology, Huashan Hospital Fudan University, Shanghai, China
| | - Zhou Yang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Guan
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuanxin Pang
- Department of Endocrinology, Suzhou Ninth People's Hospital Affiliated to Soochow University, Suzhou, China.
| | - Linkun Zhong
- Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, Guangdong Province, China.
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Jun Xiang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
15
|
Gdowicz-Kłosok A, Krześniak M, Łasut-Szyszka B, Butkiewicz D, Rusin M. Antibacterial Activity of the p53 Tumor Suppressor Protein-How Strong Is the Evidence? Int J Mol Sci 2025; 26:4416. [PMID: 40362653 PMCID: PMC12072856 DOI: 10.3390/ijms26094416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
The p53 tumor suppressor is best known for controlling the cell cycle, apoptosis, DNA repair, and metabolism, but it also regulates immunity and is able to impede the live cycle of viruses. For this reason, these infectious agents encode proteins which inactivate p53. However, what is less known is that p53 can also be inactivated by human pathogenic bacteria. It is probably not due to collateral damage, but specific targeting, because p53 could interfere with their multiplication. The mechanisms of the antibacterial activity of p53 are poorly known. However, they can be inferred from the results of high-throughput studies, which have identified more than a thousand p53-activated genes. As it turns out, many of these genes code proteins which have proven or plausible antibacterial functions like the efficient detection of bacteria by pattern recognition receptors, the induction of pro-inflammatory pyroptosis, the recruitment of immune cells, direct bactericidal activity, and the presentation of bacterial metabolites to lymphocytes. Probably there are more antibacterial, p53-regulated functions which were overlooked because laboratory animals are kept in sterile conditions. In this review, we present the outlines of some intriguing antibacterial mechanisms of p53 which await further exploration. Definitely, this area of research deserves more attention, especially in light of the appearance of antibiotic-resistant bacterial strains.
Collapse
Affiliation(s)
| | | | | | | | - Marek Rusin
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-101 Gliwice, Poland; (A.G.-K.); (M.K.); (B.Ł.-S.); (D.B.)
| |
Collapse
|
16
|
Lin Y, Zhang Y, Huang T, Chen J, Li G, Zhang B, Xu L, Wang K, He H, Chen H, Liu D, Guo S, He X, Lan P. Arginine Deprivation Induces Quiescence and Confers Vulnerability to Ferroptosis in Colorectal Cancer. Cancer Res 2025; 85:1663-1679. [PMID: 39992728 PMCID: PMC12046318 DOI: 10.1158/0008-5472.can-24-1940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 11/21/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
Metabolic reprogramming is a hallmark of cancer. Rewiring of amino acid metabolic processes provides the basis for amino acid deprivation therapies. In this study, we found that arginine biosynthesis is limited in colorectal cancer because of the deficiency of ornithine transcarbamylase. Accordingly, colorectal cancer cells met the demand for arginine by increasing external uptake. The addiction to environmental arginine resulted in the susceptibility of colorectal cancer to arginine deprivation, which dramatically decreased proliferation in colorectal cancer cells and promoted these cells to enter a reversible quiescence state. Arginine deprivation induced quiescence by activating the AMPK-p53-p21 pathway. RNA sequencing data indicated that colorectal cancer cells may be vulnerable to ferroptosis during arginine deprivation and the combination of ferroptosis inducers and arginine deprivation strongly impeded tumor growth in vivo. These findings suggest that dietary modification combined with ferroptosis induction could be a potential therapeutic strategy for colorectal cancer. Significance: Colorectal cancer dependency on arginine uptake creates a metabolic vulnerability to arginine deficiency that causes cell cycle arrest and ferroptosis sensitivity, highlighting arginine deprivation plus ferroptosis induction as a promising treatment.
Collapse
Affiliation(s)
- Yanyun Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanhong Zhang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianze Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junguo Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guanman Li
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bin Zhang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Xu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Anaesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Danling Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuang Guo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaosheng He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping Lan
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| |
Collapse
|
17
|
Mcneil TR, Sikder S, Dalal Y. Cancer cells' chamber of secrets: the link between micronuclei, chromothripsis and malignancy. Open Biol 2025; 15:240388. [PMID: 40359993 DOI: 10.1098/rsob.240388] [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: 12/03/2024] [Revised: 02/19/2025] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Micronuclei exhibit defective proteomes rendering their chromatin vulnerable to fragmentation. This fragmentation process, known as chromothripsis, promotes tumorigenesis by catalysing the activation of oncogenes and the silencing of tumor suppressors. With this role in mind, micronuclei serve as promising targets for therapeutic intervention. This review will explore recent discoveries regarding how micronuclei form, their function in catalysing chromothripsis and how chromothripsis provides a selective advantage for cancer cells.
Collapse
Affiliation(s)
| | - Sweta Sikder
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yamini Dalal
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
18
|
Yang W, Ma SC, Fang Z, Liu Y, Zhang X, Wang F, Wang C, Wang Y, Wang X, Chen W, Luo H, Yang L, Zhang S, Zeng B, Liu Z, Ou Q, Cai J, Yeung SCJ, Cheng C. TP53-centric ctDNA complements PET/CT for non-invasive assessment of pathological complete response and survival after neoadjuvant immunochemotherapy in esophageal squamous cell carcinoma: a prospective cohort study. Int J Surg 2025; 111:3256-3268. [PMID: 40146232 DOI: 10.1097/js9.0000000000002341] [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: 01/08/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Accurate assessment of pathologic complete response (pCR) after neoadjuvant immunochemotherapy (NICT) is crucial to implement active surveillance or tailor therapeutic strategies for esophageal squamous cell carcinoma (ESCC), while reliable non-invasive methods for pCR prediction are lacking. We aimed to evaluate the potential of integrating circulating tumor DNA (ctDNA) and PET/CT for predicting pCR to NICT for ESCC. METHODS A total of 123 eligible patients were enrolled, including 68 patients from our prospective clinical trial (ChiCTR2000028900) and a real-world study (NCT04822103) that formed the discovery cohort, as well as 55 patients from another clinical trial (ChiCTR2100051763) comprising the validation cohort. Blood samples for ctDNA sequencing and PET/CT metrics were collected before and after NICT. RESULTS The ctDNA status and PET/CT parameters at the post-NICT stage rather than the pre-NICT stage significantly differentiated pCR from non-pCR patients. ctDNA and PET/CT synergistically enhanced the prediction of pCR from perspectives of sensitivity and specificity, respectively. The model integrating ctDNA concentration and mean standardized uptake value (SUVmean) demonstrated area under curves (AUCs) of 0.860 in the discovery cohort and 0.798 in the validation cohort for pCR prediction and stratified patients into high- and low-risk groups with differential survival prospects. The key gene modules converged on TP53 as the core mutation for pCR prediction, among which those located in the exon regions contributed the most to its predictive capacity. The model constructed based on TP53 mutation and SUVmean differentiated pCR from non-pCR with comparable performance to the model based on PET/CT and the overall ctDNA concentration. CONCLUSION The combination of post-treatment TP53 -centric ctDNA and PET/CT synergistically enhances the prediction of pCR following NICT in ESCC patients, indicating the potential to inform clinical decision-making for these patients.
Collapse
Affiliation(s)
- Weixiong Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Si-Cong Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zengli Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yao Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fang Wang
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chenxuan Wang
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Yuze Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoyan Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenfang Chen
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Luo
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lingling Yang
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Shuishen Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bo Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhenguo Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiuxiang Ou
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Junchao Cai
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chao Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Zhu Y, Chen Z, Niu K, Li M, Deng Y, Zhang J, Wei D, Wang J, Zhao Y. USP33 Regulates DNA Damage Response and Carcinogenesis Through Deubiquitylating and Stabilising p53. Cell Prolif 2025; 58:e13793. [PMID: 39694539 PMCID: PMC12099211 DOI: 10.1111/cpr.13793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
The de-ubiquitinase USP33 has been shown to possess either tumour-promoting or inhibitory effect on human cancer cells. However, all these findings are mainly based on in vitro cell culture models, and the in vivo evidence, which is more plausible to digest the functional role of USP33 in carcinogenic process, is still lacking. Here, we demonstrate that USP33 modulates DNA damage responses including cell cycle arrest and apoptosis induction through associating with p53. It directly interacts with p53 to mediate its de-ubiquitination and further stabilisation under DNA damage condition. Depletion of USP33 induces an enhanced level of p53 ubiquitination, which de-stabilises p53 protein leading to impaired DNA damage responses. Furthermore, USP33 silencing shows either promoted or inhibited effect on cell proliferation in human cancer cells with p53 WT and mutant background, respectively. Consistently, mice with hepatocyte-specific USP33 knockout are more sensitive to nitrosodiethylamine (DEN)-induced hepatocarcinogenesis compared to wild type mice. Thus, our in vitro and in vivo evidences illustrate that USP33 possesses anti-tumour activity via regulating p53 stability and activity.
Collapse
Affiliation(s)
- Yuqi Zhu
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zixiang Chen
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Kaifeng Niu
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Mengge Li
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yuchun Deng
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ji Zhang
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Di Wei
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Jiaqi Wang
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - YongLiang Zhao
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
20
|
Wen Q, Liu J, Hu J, Kou KI, Li H, Zhang J, Zhang R, Zhong S, Huang R. Molecular mechanisms underlying the anti-Colon Cancer effects of Caulerpa lentillifera polysaccharides (CLP). Int J Biol Macromol 2025; 308:142594. [PMID: 40157667 DOI: 10.1016/j.ijbiomac.2025.142594] [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: 12/18/2024] [Revised: 03/15/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Colon cancer (CC) ranks is the second leading cause of cancer-related deaths globally. Despite chemotherapy being a primary treatment its effectiveness significantly declines in advanced in stage. Emerging evidence suggests that dietary components particularly polysaccharides, play a role in CC progression. This study employed multi-omics and network pharmacology to elucidate the mechanisms underlying the apoptotic effects of Caulerpa lentillifera polysaccharide (CLP) in CC, validated through in vitro and in vivo experiments. Transcriptomics and network pharmacology analysis identified the p53/Bax/Caspase-3 pathway as a key regulatory axis. Further targeted analysis of amino acid metabolism revealed that CLP significantly decreased intracellular aspartate (Asp) levels. Additionally, reactive oxygen species (ROS) accumulation was detected in cells. CLP treatment reduced Asp content, leading to ROS accumulation, which activated the p53/Bax/Caspase-3 pathway, triggering apoptosis. In vivo, CLP effectively inhibited tumor growth in BALB/c mice bearing CT26 colon cancer cells. These findings suggest that CLP exerts anti-colon cancer effects by modulating amino acid metabolism and inducing apoptosis via the p53/Bax/Caspase-3 axis, providing a promising therapeutic strategy for CC.
Collapse
Affiliation(s)
- Qinghua Wen
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jun Liu
- Laboratory of Pathogenic Biology, Guangdong Medical University, Zhanjiang 524023, China
| | - Jiaheng Hu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Kit Ian Kou
- Department of Mathematics, Faculty of Science and Technology, University of Macau, Macao
| | - Haichou Li
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaojiao Zhang
- College of Food and Health, Zhejiang A& F University, Hangzhou 311300, China
| | - Rongxin Zhang
- Department of Colorectal Surgery, Sun Yatsen University Cancer center, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, Guangzhou 510060, China
| | - Saiyi Zhong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; Shenzhen Research Institute, Guangdong Ocean University, Shenzhen 518108, China.
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
21
|
Li L, Li G, Zhai W. Single-cell transcriptomic analysis reveals efferocytosis signature predicting immunotherapy response in hepatocellular carcinoma. Dig Liver Dis 2025; 57:611-623. [PMID: 39904693 DOI: 10.1016/j.dld.2025.01.196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/25/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a substantial global health challenge owing to its high mortality rate and limited therapeutic options. We aimed to develop an efferocytosis-related gene signature (ER.Sig) and conduct a transcriptomic analysis to predict the prognosis and immunotherapeutic responses of patients with HCC. METHODS Single-cell RNA sequencing data and bulk RNA sequencing data were obtained from public databases. Based on single-sample gene set enrichment analysis and Weighted Gene Co-expression Network analyses, efferocytosis-related genes (ERGs) were selected at both the single-cell and bulk transcriptome levels. A machine-learning framework employing ten different algorithms was used to develop the ER.Sig. Subsequently, a multi-omics approach (encompassing genomic analysis, single-cell transcriptomics, and bulk transcriptomics) was employed to thoroughly elucidate the prognostic signatures. RESULTS Analysis of the HCC single-cell transcriptomes revealed significant efferocytotic activity in macrophages, endothelial cells, and fibroblasts within the HCC microenvironment. We then constructed a weighted co-expression network and identified six modules, among which the brown module (168 genes) was most highly correlated with the efferocytosis score (cor = 0.84). Using the univariate Cox regression analysis, 33 prognostic ERGs were identified. Subsequently, a predictive model was constructed using 10 machine-learning algorithms, with the random survival forest model showing the highest predictive performance. The final model, ER.Sig, comprised nine genes and demonstrated robust prognostic capabilities across multiple datasets. High-risk patients exhibited greater intratumoral heterogeneity and higher TP53 mutation frequencies than did low-risk patients. Immune landscape analysis revealed that compared with high-risk patients, low-risk patients exhibited a more favorable immune environment, characterized by higher proportions of CD8+ T and B cells, tumor microenvironment score, immunophenoscore, and lower Tumor Immune Dysfunction and Exclusion scores, indicating better responses to immunotherapy. Additionally, an examination of an independent immunotherapy cohort (IMvigor210) demonstrated that low-risk patients exhibited more favorable responses to immunotherapy and improved prognoses than did their high-risk counterparts. CONCLUSIONS The developed ER.Sig effectively predicted the prognosis of patients with HCC and revealed significant differences in tumor biology and treatment responses between the risk groups.
Collapse
Affiliation(s)
- Longhu Li
- Department of Intervention, Linfen Central Hospital, Linfen, PR China.
| | - Guangyao Li
- Department of Intervention, Linfen Central Hospital, Linfen, PR China
| | - Wangfeng Zhai
- Department of Intervention, Linfen Central Hospital, Linfen, PR China
| |
Collapse
|
22
|
Xia Y, Su M, Ye Z, Du F, Wang X, Guan D, Zhang X, Rao Z, Ning P. An epigenetic regulator synergizes with alphavirus-mediated gene therapy via biomimetic delivery for enhanced cancer therapy. Trends Biotechnol 2025; 43:1196-1214. [PMID: 39955233 DOI: 10.1016/j.tibtech.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 02/17/2025]
Abstract
Gene therapy is promising for treating genetic disorders, but faces challenges in treating cancer due to the intricate genetic and immunosuppressive landscape of this disease. Here, we describe a technology combining alphavirus-based gene therapy with an epigenetic regulator via pyroptosis and immune checkpoints to address these challenges. A filamentous actin-mimicking liposomal delivery system, with high fusion efficiency, was developed that encapsulates the Semliki Forest virus (pSFV) DNA vector to deliver p53 and PDL1 scFv DNA, bypassing traditional endocytic barriers to deliver genes with high efficiency via membrane fusion. To enhance this combined therapy, the DNA methyltransferase inhibitor decitabine (DAC) was used to increase Gasdermin E (GSDME) expression, converting apoptosis to pyroptosis. This approach kills apoptosis-resistant tumor cells, and also promotes T cell infiltration and activation, facilitating an anti-PDL1 therapy and the systemic antitumor immune response. This multifaceted therapeutic strategy combines gene therapy with epigenetic regulation to significantly improve immune checkpoint therapy (ICT) effectiveness, offering a robust potential as a transformative cancer treatment.
Collapse
Affiliation(s)
- Yuqiong Xia
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Maozhi Su
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Zixuan Ye
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Fuyu Du
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Xinruo Wang
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Dashan Guan
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Xianghan Zhang
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China; Guangzhou Institute of Technology, Xidian University, Guangzhou, Guangdong, 510555, PR China
| | - Zhiping Rao
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China.
| | - Pengbo Ning
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China.
| |
Collapse
|
23
|
Connolly EA, Boye K, Bonvalot S, Kratz CP, Leithner A, Malkin D, Messiou C, Miah AB, Pantziarka P, Timmermann B, van der Graaf WT, Thomas DM, Stacchiotti S. Genetic predisposition in sarcomas: clinical implications and management. EClinicalMedicine 2025; 83:103203. [PMID: 40291347 PMCID: PMC12032185 DOI: 10.1016/j.eclinm.2025.103203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 03/14/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Recent studies indicate up to 20% of sarcomas may be associated with predisposition genes, and this number will probably increase as genetic testing becomes more available. Evidence on the management of patients with sarcoma and genetic predisposition remains, however, scarce. This review compiles available research on genetic predisposition syndromes associated with sarcoma and sarcoma treatment within such syndromes, addressing key gaps in knowledge. We explore the current evidence on how genetic predisposition may influence treatment decisions and clinical management, focusing on surgery, radiotherapy, systemic treatment, and surveillance. Evidence-based recommendations are currently not available for most syndromes, and we have therefore included pragmatic advice for clinicians. Unanswered questions and unmet needs are also identified, underscoring the importance of multidisciplinary input from specialists such as geneticists, radiologists, surgeons and oncologists. The review stresses the need for future research to improve clinical outcomes for patients with sarcoma and genetic predisposition. Funding No funding has been provided for this work.
Collapse
Affiliation(s)
- Elizabeth A. Connolly
- Department of Medical Oncology, Chris O’Brien Lifehouse, Sydney, Australia
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Kjetil Boye
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sylvie Bonvalot
- Department of Surgery, Institut Curie, Comprehensive Cancer Center, Paris, France
| | - Christian P. Kratz
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Andreas Leithner
- Department of Orthopedics and Trauma, Medical University of Graz, Graz, Austria
| | - David Malkin
- Division of Haematology-Oncology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Christina Messiou
- Sarcoma Unit, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
- The Institute of Cancer Research, London, United Kingdom
| | - Aisha B. Miah
- Sarcoma Unit, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
- The Institute of Cancer Research, London, United Kingdom
| | - Pan Pantziarka
- Anticancer Fund, Meise, Belgium
- George Pantziarka TP53 Trust, London, United Kingdom
| | - Beate Timmermann
- Department of Particle Therapy, University Hospital Essen, West German Proton Therapy Centre Essen (WPE), Essen, Germany
| | - Winette T.A. van der Graaf
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - David M. Thomas
- Garvan Institute of Medical Research, Sydney, Australia
- Centre for Molecular Oncology, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
24
|
Wang B, Huang K, Xiao J, Tao Y, Luo J, Wu Y, Zhou S, He Y, He L. Ursolic acid induces apoptosis in nasopharyngeal carcinoma cells through the P53 signaling pathway: a network pharmacology and experimental validation study. Med Oncol 2025; 42:189. [PMID: 40310511 DOI: 10.1007/s12032-025-02749-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/28/2025] [Indexed: 05/02/2025]
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant neoplasm that is highly prevalent in East Asia and presents significant therapeutic challenges due to limited treatment options and severe adverse effects. Ursolic acid (UA) is a pentacyclic triterpenoid with anticancer activity in various tumors; however, its mechanism of action in NPC remains unclear. This study integrated network pharmacology with experimental validation to elucidate the molecular mechanism underlying the effect of UA against NPC. Screening of a network pharmacology database identified 39 targets common to UA and NPC, among which P53, STAT3, Bcl-2, IL1B, and CASP3 showed high node degrees in the protein-protein interaction network. Gene Ontology analysis revealed that these targets were primarily enriched in stress response and apoptosis regulation, whereas Kyoto Encyclopedia of Genes and Genomes analysis indicated significant enrichment in the P53 signaling and apoptosis pathways. UA dose-dependently inhibited the proliferation of the NPC cell lines S18 and S26 (p < 0.01), and induced apoptosis, as demonstrated by Annexin V-FITC/PI double fluorescence staining and confirmed by Hoechst 33,342 staining showing nuclear condensation. UA also caused mitochondrial membrane depolarization, as indicated by JC-1 staining. Western blot analysis showed significant upregulation of P53 and the pro-apoptotic protein BAX (p < 0.01), and downregulation of the anti-apoptotic protein Bcl-2 (p < 0.01) following UA treatment. This study is the first to show that UA induces apoptosis in NPC cells by activating the P53 signaling pathway using network pharmacology and experimental validation.
Collapse
Affiliation(s)
- Binya Wang
- Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Keying Huang
- Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jiale Xiao
- Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yangyang Tao
- Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jingjing Luo
- Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yonghui Wu
- Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Sainan Zhou
- Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yingchun He
- Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Lan He
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China.
- Hunan Provincial Engineering and Technological Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases With Chinese Medicine and Protecting Visual Function, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
25
|
Wei Q, Li C, Tang Y, Bai J, Li W, Liu J, Su Z, Cheng X. Mechanistic Role of the Mdm2/MdmX Lid Domain in Regulating Their Interactions with p53. Biomolecules 2025; 15:642. [PMID: 40427535 PMCID: PMC12109074 DOI: 10.3390/biom15050642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025] Open
Abstract
p53 functions as a critical guardian of the genome, orchestrating tumor suppression pathways and ensuring the integrity of chromosomal stability. Mdm2 and MdmX, homologous proteins, serve as negative feedback regulators of p53. In approximately half of tumor cases, overexpression of Mdm2/MdmX results in the inhibition of p53 activity. Current research focuses on designing Mdm2 and MdmX inhibitors based on the structure of lidless N-terminal forms of these proteins. However, growing evidence suggests that the lid of Mdm2 and MdmX plays a key role in the selective binding of p53 and inhibitors. Therefore, targeting the lid in the screening and design of Mdm2/MdmX inhibitors may offer a novel strategy for developing anti-cancer drugs. This review examines the impact of the Mdm2/MdmX lid on ligand binding, providing valuable insights for future research and guiding new approaches to the screening and design of innovative anti-cancer therapeutics.
Collapse
Affiliation(s)
- Qiuyin Wei
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Q.W.); (C.L.); (Y.T.); (J.B.); (W.L.); (J.L.)
| | - Chenqi Li
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Q.W.); (C.L.); (Y.T.); (J.B.); (W.L.); (J.L.)
| | - Yibing Tang
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Q.W.); (C.L.); (Y.T.); (J.B.); (W.L.); (J.L.)
| | - Jinping Bai
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Q.W.); (C.L.); (Y.T.); (J.B.); (W.L.); (J.L.)
| | - Wang Li
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Q.W.); (C.L.); (Y.T.); (J.B.); (W.L.); (J.L.)
| | - Jidong Liu
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Q.W.); (C.L.); (Y.T.); (J.B.); (W.L.); (J.L.)
| | - Zhengding Su
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China;
| | - Xiyao Cheng
- Institute of Modern Fermentation Engineering and Future Foods, School of Light Industry and Food Engineering, Guangxi University, No. 100, Daxuedong Road, Nanning 530004, China; (Q.W.); (C.L.); (Y.T.); (J.B.); (W.L.); (J.L.)
| |
Collapse
|
26
|
Yan Q, Wong W, Lei J, Liang D, Yang J, Gong L, Berardi R, Dai S, Wang J. LNCAROD was stabilized through N 6-methyladenosine methylation and exerted its anticancer effects in lung squamous cell carcinoma by inhibiting SIRT1 activity via CCAR2. Transl Lung Cancer Res 2025; 14:1351-1370. [PMID: 40386722 PMCID: PMC12082203 DOI: 10.21037/tlcr-2025-267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025]
Abstract
Background Lung squamous cell carcinoma (LUSC), a deadly malignant tumor, is highly prevalent worldwide. Accumulating evidence indicates that long-chain noncoding RNAs play crucial regulatory roles in the occurrence and progression of LUSC. LNCAROD regulates the proliferation, migration, and invasion of cells by upregulating SERPINE1 expression in lung adenocarcinoma (LUAD). However, the functional mechanism of LNCAROD action in LUSC remains unclear. The aim of this study was to investigate the regulatory function and mechanism of LNCAROD action in the development of LUSC. Methods Using quantitative polymerase chain reaction (qPCR) detection, we determined the expression of LNCAROD in LUSC tissues and cell lines. Cell Counting Kit-8 (CCK-8), EdU (5-ethynyl-2'-deoxyuridine), JC-1 mitochondrial membrane potential, flow cytometry, colony formation, scratch healing, and Transwell assays were conducted, and cell proliferation, migration, and invasion, as well as physiological changes were assessed. The tumorigenicity of LUSC cells was analyzed by in vitro tumor formation in nude mice. Molecular interactions were verified via Western blotting, RNA-protein pull-down assay, RNA binding protein immunoprecipitation (RIP), N6-methyladenosine (m6A)-RIP, and coimmunoprecipitation (Co-IP) analyses. Results LNCAROD was specifically and highly expressed in LUSC cells and tissues. LNCAROD expression was mediated by IGF2BP2 m6A methylation, which, along with CCAR2, inhibited SIRTI1's acetylation activity. This further induced p53 protein acetylation and promoted the mitochondrial apoptosis of LUSC cells, thereby inhibiting cell proliferation, migration, and invasion. Conclusions LNCAROD is specifically highly expressed in LUSC cells and tissues and may be a tumor-suppressor gene. The findings contribute to a deeper understanding of the function of LNCAROD in LUSC, and it may serve as a potential prognostic marker for personalized medical diagnosis in clinical practice.
Collapse
Affiliation(s)
- Qihang Yan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wingshing Wong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Guangzhou, China
| | - Jinsong Lei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dachuan Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jie Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Gong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | - Shuqin Dai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Junye Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
27
|
Huang S, Yin H. Exploring the Molecular Mechanism and Role of Glutathione S-Transferase P in Prostate Cancer. Biomedicines 2025; 13:1051. [PMID: 40426879 PMCID: PMC12109251 DOI: 10.3390/biomedicines13051051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Aims: To investigate the effect of Glutathione metabolism in prostate cancer pathogenesis. Background: There is growing evidence that Glutathione metabolism plays an important role in prostate cancer, with genes encoding key enzymes in this pathway potentially serving as diagnostic or prognostic biomarkers. Objective: To explore whether there is a causal relationship between key enzymes in the Glutathione metabolism and prostate cancer, and to further investigate the molecular mechanisms and roles of the genes encoding their proteins in relation to prostate cancer. Method: Transcriptomic datasets from the Gene Expression Omnibus (GEO) database were analyzed to identify differentially expressed genes (DEGs) and enriched pathways in prostate cancer versus normal tissues. Two-sample bidirectional Mendelian randomization (MR) was employed to assess causal relationships between Glutathione metabolic enzymes (exposure) and prostate cancer risk (outcome). Immune infiltration analysis and LASSO regression were performed to construct a diagnostic model. Single-cell RNA sequencing (scRNA-seq) data were utilized to elucidate cell-type-specific expression patterns and functional associations of target genes. Result: The results of two-sample bidirectional MR showed that Glutathione S-transferase P (GSTP) in Glutathione metabolism could reduce the risk of prostate cancer. The Glutathione S-transferase Pi-1 (GSTP1) gene was lowly expressed in prostate cancer and was able to diagnose prostate cancer more accurately. Single-cell analysis showed that the high expression of GSTP1 in prostate cancer epithelial cells was closely associated with the upregulation of the P53 pathway and apoptosis. Conclusions: Our study reveals that GSTP in Glutathione metabolism reduces the risk of prostate cancer and further analyzes the genetic association and mechanism of action between GSTP1 and prostate cancer.
Collapse
Affiliation(s)
- Shan Huang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China;
- Institute of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Hang Yin
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China;
- Institute of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
28
|
Ji Z, Zhang Y, Hu G, Hong S, Su Z, Zhang Q, Wang L, Wang T, Yu S, Bu Q, Yuan F, Zhu X, Jia G. Hexavalent chromium and cellular senescence: A comprehensive analysis from chromate-exposed occupational population and chromate-inhaled mouse model. JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138387. [PMID: 40286659 DOI: 10.1016/j.jhazmat.2025.138387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Cellular senescence may predominantly drive the progression of early subclinical injury under conditions of low-dose, long-term occupational exposure. However, previous research has largely overlooked the cellular senescence induced by hexavalent chromium [Cr(VI)]. To bridge the gap, 304 workers from a chromate facility were enrolled, and a mouse model was used to confirm the effects of Cr(VI) on cellular senescence. A 2.7-fold increase in blood Cr was related to the changes of p53 [23.19 (13.06, 34.23)%], serum α-Klotho [11.45 (6.13, 17.04)%], adipsin [-14.11(-22.16, -5.24)%], leptin [-4.32(-6.99, -1.58)%] and resistin [-3.29(-5.54, -0.98)%]. There were significant correlations of blood Cr with DNA methylation of ELOVL2 and hTERT genes. Furthermore, methylation at hTERT Pos1, Pos2, Pos6, and Pos8 significantly mediated the relationship between blood Cr and p53. In the mouse model, we observed significantly higher mRNA expression levels of key genes in the p53/p21 and Rb/p16 pathways and senescence-associated β-galactosidase positive cell ratio in the exposed group. In conclusion, we found that p53 in human peripheral blood cells serves as a Cr(VI)-induced senescence biomarker, with α-Klotho upregulation and adipokines (adipsin, leptin, and resistin) downregulation indicating compensatory responses, as well as hTERT methylation partially mediating Cr(VI)-senescence association.
Collapse
Affiliation(s)
- Zhiqiang Ji
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, PR China
| | - Yali Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, PR China
| | - Guiping Hu
- School of Engineering Medicine and Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, PR China
| | - Shiyi Hong
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, PR China
| | - Zekang Su
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, PR China
| | - Qiaojian Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, PR China
| | - Li Wang
- Department of Toxicology, School of Public Health, Baotou Medical College, Baotou, Inner Mongolia 014040, PR China
| | - Tiancheng Wang
- Department of Clinical Laboratory, Third Hospital of Peking University, Beijing 100191, PR China
| | - Shanfa Yu
- Henan Institute for Occupational Medicine, Zhengzhou, Henan 450052, PR China
| | - Qian Bu
- The Disease Control and Prevention Center of Tongnan District, Chongqing 402660, PR China
| | - Fang Yuan
- Department of Occupational Health and Radiological Health, Chongqing Center for Disease Control and Prevention, Chongqing 400042, PR China
| | - Xiaojun Zhu
- National Center for Occupational Safety and Health, Beijing 102308, PR China
| | - Guang Jia
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing 100191, PR China.
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
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
|
32
|
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
|
33
|
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
|
34
|
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
|
35
|
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
|
36
|
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
|
37
|
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
|
38
|
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
|
39
|
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] [Download PDF] [Figures] [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
|
40
|
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
|
41
|
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
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
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
|
46
|
Zhou Y, Zhang H, Yan H, Han P, Zhang J, Liu Y. Deciphering the Role of Oxidative Stress in Male Infertility: Insights from Reactive Oxygen Species to Antioxidant Therapeutics. FRONT BIOSCI-LANDMRK 2025; 30:27046. [PMID: 40302329 DOI: 10.31083/fbl27046] [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: 10/20/2024] [Revised: 12/25/2024] [Accepted: 01/10/2025] [Indexed: 05/02/2025]
Abstract
Male infertility represents a major health concern, accounting for approximately 50% of all infertility cases in couples. This condition arises from multiple etiologies, with oxidative stress gaining increasing attention in recent studies. During the final stages of sperm maturation, the majority of the cytoplasm is discarded, leaving sperm with a diminished antioxidant defense system, which makes them highly susceptible to the detrimental effects of reactive oxygen species (ROS). ROS can be generated from both intrinsic and extrinsic sources. Intrinsically, ROS are primarily produced by mitochondrial activity, while extrinsic factors include alcohol consumption, smoking, circadian rhythm disruption, gut microbiota imbalance, and leukocyte infiltration. Excessive ROS production leads to DNA damage, apoptosis, and epigenetic modifications in sperm, ultimately impairing sperm motility and contributing to infertility. This review provides a comprehensive examination of ROS sources and examines the mechanisms by which ROS induce sperm damage. Furthermore, it explores the therapeutic potential of antioxidants in mitigating oxidative stress and improving sperm quality.
Collapse
Affiliation(s)
- Ye Zhou
- Department of Reproductive Medicine, Zhaotong Hospital of Traditional Chinese Medicine, 657000 Zhaotong, Yunnan, China
| | - Hengyan Zhang
- Department of Dermatology, Zhaotong Hospital of Traditional Chinese Medicine, 657000 Zhaotong, Yunnan, China
| | - Heguo Yan
- Department of Clinical Medicine, Yunnan University of Chinese Medicine, 650500 Kunming, Yunnan, China
- Department of Endocrinology, Zhaotong Hospital of Traditional Chinese Medicine, 657000 Zhaotong, Yunnan, China
| | - Pingxing Han
- Department of Reproductive Medicine, Zhaotong Hospital of Traditional Chinese Medicine, 657000 Zhaotong, Yunnan, China
| | - Jing Zhang
- Department of Reproductive Medicine, Zhaotong Hospital of Traditional Chinese Medicine, 657000 Zhaotong, Yunnan, China
| | - Yangwen Liu
- Department of Endocrinology, Zhaotong Hospital of Traditional Chinese Medicine, 657000 Zhaotong, Yunnan, China
| |
Collapse
|
47
|
Ma J, Cai Y, Lu Y, Fang X. Analysis and assessment of ferroptosis-related gene signatures and prognostic risk models in skin cutaneous melanoma. Transl Cancer Res 2025; 14:1857-1873. [PMID: 40224981 PMCID: PMC11985187 DOI: 10.21037/tcr-24-1506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 01/14/2025] [Indexed: 04/15/2025]
Abstract
Background The occurrence and development of skin cutaneous melanoma (SKCM) are significantly influenced by ferroptosis, a sort of regulated cell death characterized by iron deposition and lipid peroxidation. Although positive strides have been achieved in the present management of SKCM, it is still unknown exactly how ferroptosis occurs in this condition. We aimed to determine the role of prognostically relevant ferroptosis-related genes (PR-FRGs) in SKCM development and prognosis. Methods The training group was created using combined transcriptomic RNA data acquired from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. The dataset GSE19234 was acquired from the Gene Expression Omnibus (GEO) database as a validation group. Differentially expressed ferroptosis-related genes (DE-FRGs) were obtained from the training group, of which 103 showed up-regulation and 77 showed down-regulation. Then, 12 PR-FRGs were identified by the protein-protein interaction (PPI) network and Cox regression analysis, and prognostic risk models and nomograms were constructed. The risk model was validated using a validation group, and the prognostic value of the risk model was analyzed. Finally, immunohistochemical data were obtained from the Human Protein Atlas (HPA) website to validate the PR-FRGs. Results Twelve PR-FRGs were identified. A prognostic risk model was built using PR-FRGs, and patients in the training and validation groups were classified as high or low risk based on the risk model. The outcomes demonstrated that the prognosis was better for the low-risk group. Prognostic value analysis showed that the prognostic risk model could accurately predict the patients' overall survival (OS), was superior to clinical traits such as age, gender, and tumor stage in predicting ability, and could be used as an independent predictor. Meanwhile, the nomogram constructed based on PR-FRGs can effectively predict the prognosis of SKCM patients. Finally, PR-FRGs were validated in the HPA database. Conclusions Ferroptosis affects the prognosis of SKCM patients. Prognostic risk model and nomogram constructed based on 12 PR-FRGs demonstrated significant advantages in predicting the prognosis of SKCM patients. This will help in the identification and prognostic prediction of SKCM and in the discovery of new individualized treatment modalities.
Collapse
Affiliation(s)
- Jianchao Ma
- Department of Orthopedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yang Cai
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Youqi Lu
- Department of Orthopedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xu Fang
- Department of Orthopedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
48
|
Ding M, Huo K, Chen X, Wang W, Xiang Z, Song Y, Chen P, Liu L. The role of non-coding RNA in ferroptosis of liver cancer and its impact on lipid peroxidation. Front Immunol 2025; 16:1555518. [PMID: 40207231 PMCID: PMC11979700 DOI: 10.3389/fimmu.2025.1555518] [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: 01/04/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
Ferroptosis is an iron-dependent programmed death caused by the imbalance of lipid peroxides in cells. Unlike apoptosis, autophagy and necrosis, ferroptosis is mainly induced by the small molecule compound erastin. The main characteristics of ferroptosis were glutathione (GSH) depletion, inactivation of glutathione peroxidase 4 (GPX4) and reactive oxygen species (ROS) promoting lipid peroxidation. Eventually, the imbalance of lipid peroxidation regulation in cells leads to ferroptosis. The lipid metabolic pathway ultimately contributes to ferroptosis through the production of lipid peroxides. In addition, other cellular metabolic pathways can also regulate ferroptosis, such as the antioxidant metabolic pathway, which inhibits ferroptosis by clearing lipid peroxides and reducing cell membrane damage. Long non-coding RNAs (lncRNAs) are non-coding transcripts more than 200 nucleotides in length and are a less classified group of RNA transcripts that are associated with tumorigenesis and metastasis and are more tissue or cell type specific than protein-coding genes. Studies on the molecular profile of lncRNAs in plasma samples from liver cancer patients show that differentially expressed lncRNAs are mainly concentrated in biological functions related to tumorigenesis, such as cell metastasis, immune response and metabolic regulation. With different biological functions in physiological and pathological environments, the specific expression patterns of lncRNAs coordinate cell state, development, differentiation, and disease.
Collapse
Affiliation(s)
- Minglu Ding
- Graduate Student Department, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Keyuan Huo
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xiaojie Chen
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Wanyao Wang
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Zihan Xiang
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Yidan Song
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Peijian Chen
- College of Life Science, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Lantao Liu
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| |
Collapse
|
49
|
Otero-Mateo M, Estrany Jr F, Arcas-Márquez S, Moya-Borrego L, Castellano G, Castany M, Alemany R, Fillat C. KPC pancreatic cancer cells are a novel immunocompetent murine model supporting human adenovirus replication and tumor oncolysis. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200928. [PMID: 39877727 PMCID: PMC11773232 DOI: 10.1016/j.omton.2024.200928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/31/2025]
Abstract
Oncolytic adenoviral therapy is a promising approach for pancreatic cancer treatment. However, the limited capacity of murine cells to produce infectious viral progeny precludes the full evaluation of the virotherapy in a suitable immunocompetent mouse model. Here, we report that the murine KPC-I cell line, established from pancreatic tumors developed in LSL-K r as G12D ; LSL-T r p53 R172H ; Pdx-Cre mice, is susceptible to adenoviral replication and generates a progeny of infective virions similar to those from infected human A549 cells. A comparative study with the semipermissive murine CMT64.6 cells reveals that adenoviral infection of KPC-I cells substantially increases the release of infective particles, with a correlating enhanced susceptibility to adenovirus-induced autophagy. Remarkably, systemic delivery of the oncolytic adenovirus AdNuPARE1A in athymic mice bearing KPC-I tumors results in significant inhibition of tumor growth. Moreover, KPC-I tumors in immunocompetent mice with intratumoral administration of AdNuPARE1A or ICOVIR15kDelE3 display significant antitumoral effects, with evidence of adenoviral replication. Collectively, our data show that KPC-I cells are permissive to human oncolytic adenovirus replication, rendering KPC-I syngeneic tumors an interesting model to evaluate the multifaceted antitumor activities of oncolytic adenovirus.
Collapse
Affiliation(s)
- Marc Otero-Mateo
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Francesc Estrany Jr
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Programa de Biomedicina. Universitat de Barcelona, 08036 Barcelona, Spain
| | - Sabrina Arcas-Márquez
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Laura Moya-Borrego
- Cancer Immunotherapy Group, Oncobell and iProCURE Programs, IDIBELL-Institut Català d’Oncologia, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Giancarlo Castellano
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Miquel Castany
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Ramon Alemany
- Cancer Immunotherapy Group, Oncobell and iProCURE Programs, IDIBELL-Institut Català d’Oncologia, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Fillat
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08036 Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut. Universitat de Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
50
|
ZHAO SHUANG, WEN HONGYONG, WANG BAIQI, XIONG QINGLIN, LI LANXIN, CHENG AILAN. p53: A player in the tumor microenvironment. Oncol Res 2025; 33:795-810. [PMID: 40191727 PMCID: PMC11964878 DOI: 10.32604/or.2025.057317] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/09/2024] [Indexed: 04/09/2025] Open
Abstract
Approximately half of all cancers have p53 inactivating mutations, in addition to which most malignancies inactivate the p53 pathway by increasing p53 inhibitors, decreasing p53 activators, or inactivating p53 downstream targets. A growing number of researches have demonstrated that p53 can influence tumor progression through the tumor microenvironment (TME). TME is involved in the process of tumor development and metastasis and affects the clinical prognosis of patients. p53 participates in host immunity and engages in the immune landscape of the TME, but the specific mechanisms remain to be investigated. This review briefly explores the interactions between different states of p53 and TME components and their mechanisms, as well as their effects on tumor progression. To understand the progress of drug development and clinical studies related to p53 and tumor microenvironment.
Collapse
Affiliation(s)
- SHUANG ZHAO
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - HONGYONG WEN
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - BAIQI WANG
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - QINGLIN XIONG
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - LANXIN LI
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - AILAN CHENG
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|