1
|
He J, Li T, Pan X, Deng Z, Huang J, Mo X, Shen X, Qin X, Yang X, Gao M, Yang J. CD44 and αV-integrins dual-targeting bimetallic nanozymes for lung adenocarcinoma therapy via NIR-enhanced ferroptosis/apoptosis. Biomaterials 2025; 323:123407. [PMID: 40403445 DOI: 10.1016/j.biomaterials.2025.123407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/27/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025]
Abstract
Combination therapy is a promising strategy for lung adenocarcinoma (LUAD), due to the advantages of overcoming drug resistance, side effects, and tumor heterogeneity. Herein, we report a novel dual-targeting bimetallic nanozyme (MH-iRGD) consisting of nanosized manganese ferrite (MF) after encapsulating with dopamine and methacrylic anhydride to modify hyaluronic acid, followed by integrin receptor targeting peptide (HS-PEG3400-iRGD) modification for LUAD targeted therapy. Our study confirmed that MH-iRGD combined with near-infrared irradiation (NIR) possessed dramatic photothermal effects and reactive oxygen species (ROS) production and GSH depletion abilities. Importantly, MH-iRGD possessed dual-targeting capacities for LUAD cells overexpressed CD44 and αV-integrin receptors owing to hyaluronic acid coating and iRGD modification. Inhibitors of CD44 and integrins could impair the uptake of MH-iRGD in LUAD cells. Moreover, MH-iRGD + NIR displayed excellent anti-LUAD effects as a result of the production of intracellular ROS, consumption of glutathione (GSH) and mitochondrial dysfunction. Mechanistically, NIR robustly strengthened MH-iRGD-induced ferroptosis and apoptosis by down-regulating SLC7A11, GPX4, Bcl-2 levels while up-regulating Bax level. Specifically, ferroptosis and apoptosis were increased while the LUAD progression was inhibited after intravenous injection of MH-iRGD + NIR in xenograft mouse models. Taken together, our results indicate that MH-iRGD + NIR serves as a promising targeted therapy for LUAD, which broadens the applications of highly active dual-targeting bimetallic nanozymes.
Collapse
Affiliation(s)
- Jingchuan He
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China; Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Tingting Li
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China; Department of Pharmacy, The Second Affiliated Hospital of North Sichuan Medical College, Nanchong, 637100, China
| | - Xiaoqin Pan
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China
| | - Zhihua Deng
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China; Department of Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University for Nationalities, Baise, 533000, China
| | - Jifu Huang
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China
| | - Xiaocheng Mo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China; Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xiaoju Shen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China; Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xiumei Qin
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China
| | - Xingye Yang
- Department of Pharmaceutics, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China
| | - Ming Gao
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China.
| | - Jie Yang
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Drug Basic Research for Prevention and Treatment of Geriatric Diseases, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China; The Laboratory of Toxicology of Traditional Chinese Medicine, Level III Laboratory of National Administration of Traditional Chinese Medicine, School of Pharmacy, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
2
|
Shen T, Wang Y, Cheng L, Bode AM, Gao Y, Zhang S, Chen X, Luo X. Oxidative complexity: The role of ROS in the tumor environment and therapeutic implications. Bioorg Med Chem 2025; 127:118241. [PMID: 40383035 DOI: 10.1016/j.bmc.2025.118241] [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: 01/27/2025] [Revised: 05/01/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Reactive oxygen species (ROS) constitutes a group of reactive molecules that play a critical role in biological processes. Varying ROS levels have been frequently observed in cancer cells and the tumor microenvironment (TME). The role of ROS displays significant complexity in cancer development and therapy. Elevated ROS levels can induce metabolic reprogramming and promote the proliferation, invasion, and metastasis of cancer cells, resulting in cancer progression. However, excessive ROS accumulation leads to the occurrence of apoptosis, pyroptosis, necroptosis, and ferroptosis in cancer cells, which restrains tumor development. In the TME, ROS frequently promotes angiogenesis and remodels the extracellular matrix (ECM) by enhancing the differentiation of cancer-associated fibroblasts (CAFs), thereby supporting tumor growth. Concurrently, high ROS levels favour immunosuppressive cells, including M2-polarized macrophages, and regulatory T cells (Tregs), while impairing the antitumor capabilities of T cells. In the aspect of cancer therapy, it is overly simplistic to merely combine chemoradiotherapy with antioxidants as a therapeutic strategy. Instead, highlighting targeted therapies that modulate ROS is essential, given their inherent complexity. Fortunately, a variety of innovative treatments have emerged, including nanodrug delivery systems (NDDS), proteolysis-targeting chimeras (PROTAC), and adoptive cell therapy (ADT), which not only exhibit synergistic effects with immune checkpoint therapy (ICT), but also enhance the antitumor capabilities of the TME. In this paper, we elucidate the mechanism of ROS production, enumerate the role of ROS in cancer development and the TME, and discuss advancements in ROS-targeted cancer therapeutics.
Collapse
Affiliation(s)
- Tingfeng Shen
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yutong Wang
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Linmao Cheng
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Ya Gao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Shuntong Zhang
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xue Chen
- Early Clinical Trial Center, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China.
| |
Collapse
|
3
|
Xia F, Sha Y, Jin Y, Yang J, Chen C, Gong B, Liu Y, Zhao Q. Autophagy inhibition amplifies anti-tumor immunity effect of dinutuximab beta on neuroblastoma via the VEGFR/AKT/mTOR and ROS/NF-κB pathways. Int Immunopharmacol 2025; 158:114862. [PMID: 40378433 DOI: 10.1016/j.intimp.2025.114862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025]
Abstract
Dinutuximab beta has shown limited efficacy in treating high-risk neuroblastoma (NB). Combining autophagy inhibitors with immune checkpoint inhibitors (ICIs) has proven effective in many malignancies. However, the anti-tumor effects of autophagy inhibition in conjunction with anti-GD2 immunotherapy remain unknown. In this study, dinutuximab beta induces anti-proliferation and anti-EMT activity in NB cells. Dinutuximab beta also triggers autophagy in NB cells, and inhibition of the VEGFR pathway with anlotinib amplifies dinutuximab beta-induced autophagy. In addition, dinutuximab beta induces the synthesis of the chemokine CXCL9 and the infiltration of CD8+ T cells. Mechanistically, dinutuximab beta inhibits the VEGFR/AKT/mTOR and ROS/NF-κB pathways. Furthermore, autophagy inhibition by CQ enhances CXCL9 expression and anti-tumor T cell responses of single anti-GD2 therapy in vitro and in vivo. Collectively, this study suggests autophagy inhibitors may be a promising strategy for enhancing therapeutic efficacy in NB in conjunction with anti-GD2 immunotherapy.
Collapse
Affiliation(s)
- Fantong Xia
- Radiation Oncology Center, Chongqing University Cancer Hospital, College of Medicine, Chongqing University, Chongqing, China
| | - Yongliang Sha
- Department of General Surgery, Xuzhou Central Hospital, Xuzhou, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jiaxing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chong Chen
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yun Liu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
4
|
Zhou J, Shu QJ, Wang T, Huang HD, Zhang SP, Zhang J, Zheng YQ, Zhang C. Piperlongumine induces ROS accumulation to reverse resistance of 5-FU in human colorectal cancer via targeting TrxR. Eur J Pharmacol 2025; 997:177478. [PMID: 40054719 DOI: 10.1016/j.ejphar.2025.177478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Resistance is a major concern for colorectal cancer patients undergoing chemotherapy. Piperlongumine (PL) has been proven to effectively reverse drug resistance in several types of cancers; however, the mechanisms associated with the reversal effect and the targets of PL in cancer drug resistance are still unclear. In this research, the reversal effects and associated mechanisms of PL in 5-Fluorouracil (5-FU) resistance colorectal cancer were investigated both in vitro and in vivo. Our data revealed that PL acted as a ROS inducer via binding and inhibiting TrxR (IC50 around 10.17 μM). By inducing ROS accumulation, PL reversed resistance to 5-FU in HCT-8/5-FU cells (reversal ratio: 4.9-fold) and enhanced the therapeutic effects of 5-FU through the dephosphorylation of Akt in BALB/c athymic nude mice bearing HCT-8/5-FU tumor xenografts. As a ROS inducer, PL reversed resistance to 5-FU by directly promoting inhibition of Akt phosphorylation, and further inhibited 5-FU efflux and promoted cell apoptosis through affecting the Akt/Foxo3/NRF2/P-gp and Akt/Foxo3/NRF2/BAD signaling pathway.
Collapse
Affiliation(s)
- Ji Zhou
- Center for Reproductive Medicine, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Qing-Ju Shu
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Tian Wang
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Hui-Dan Huang
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Sheng-Peng Zhang
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Jing Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Yong-Qiu Zheng
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China.
| | - Chao Zhang
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China.
| |
Collapse
|
5
|
Yuan H, Liu J, Xu R, Yang K, Qu R, Liu S, Zhang Y, Xiang M. The spatiotemporal heterogeneity of reactive oxygen species in the malignant transformation of viral hepatitis to hepatocellular carcinoma: a new insight. Cell Mol Biol Lett 2025; 30:70. [PMID: 40517270 PMCID: PMC12167593 DOI: 10.1186/s11658-025-00745-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
During the transformation of viral hepatitis into hepatocellular carcinoma (HCC), oxidative stress levels increase significantly, leading to tissue damage and chronic inflammation. HCC is characterized by spatiotemporal heterogeneity, which influences oxidative stress patterns, with reactive oxygen species (ROS) as the primary representative molecules. ROS serve not only as critical biomarkers of cancer but also as potential therapeutic targets for HCC, given that their increased levels can either promote or inhibit disease progression. In this review, we systematically examine the temporal heterogeneity of ROS, emphasizing its role in different stages of HCC progression caused by viral hepatitis and in influencing cell fate. We further explore ROS spatial heterogeneity at three levels: cellular, organelle, and biomolecular. Next, we comprehensively review clinical applications and potential therapies designed to selectively modulate ROS on the basis of its spatiotemporal heterogeneity. Finally, we discuss potential future applications of novel therapies that target ROS spatiotemporal heterogeneity to prevent and manage HCC onset and progression. In conclusion, this review enhances understanding of ROS in the progression of viral hepatitis to HCC and offers insights into developing new therapeutic targets and strategies centered on ROS heterogeneity.
Collapse
MESH Headings
- Humans
- Reactive Oxygen Species/metabolism
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/virology
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/virology
- Oxidative Stress
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Animals
- Hepatitis, Viral, Human/metabolism
- Hepatitis, Viral, Human/pathology
- Hepatitis, Viral, Human/complications
- Disease Progression
Collapse
Affiliation(s)
- Huimin Yuan
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Ruochen Xu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Keshan Yang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Ruiyang Qu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Shuai Liu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
| |
Collapse
|
6
|
Bhuvaneshwari K, Harithpriya K, Ganesan K, Xu B, Ramkumar KM. Role of oxeiptosis in disease mechanisms and therapeutic opportunities. Apoptosis 2025; 30:1182-1201. [PMID: 40064755 DOI: 10.1007/s10495-025-02087-z] [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] [Accepted: 02/04/2025] [Indexed: 06/16/2025]
Abstract
Cell death is a crucial mechanism through which cells respond to damage and stress, thereby maintaining homeostasis. Cell death pathways include both caspase-dependent and caspase-independent mechanisms, such as apoptosis, necrosis, autophagy, and ferroptosis. The recent discovery of oxeiptosis identifies a unique form of ROS-mediated, caspase-independent cell death with apoptotic-like features. This process is regulated by key molecules, including KEAP1, PGAM5, and AIFM1, and is characterized by distinct molecular and morphological features. These regulators contribute to cellular integrity by activating cytoprotective genes through Nrf2 stabilization by KEAP1 and maintaining cellular homeostasis via PGAM5-mediated AIFM1 Ser116 dephosphorylation. In this review, we discuss the broad spectrum of oxeiptosis-mediated regulation in disease pathogenesis by combating ROS-induced cellular damage. Modulating oxeiptosis helps in disease management by mitigating ROS-induced cellular damage, restoring redox balance, and preventing pathological inflammation. Additionally, we highlight modulators such as natural derivatives and lncRNAs that trigger oxeiptosis in various diseases, including vitiligo, psoriasis, and multiple cancer types. Modulating oxeiptosis presents significant clinical implications by offering novel therapeutic strategies to mitigate oxidative stress, restore cellular homeostasis, and prevent inflammation-driven diseases. This review emphasizes potential therapeutic advances for conditions characterized by aberrant ROS accumulation, offering innovative avenues for clinical intervention and treatment development.
Collapse
Affiliation(s)
- K Bhuvaneshwari
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, TN, India
| | - Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, TN, India
| | - Kumar Ganesan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 3, Sassoon Road, Pokfulam, Hong Kong, 999077, China
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong, 519087, China.
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, TN, India.
| |
Collapse
|
7
|
Wang TH, Chou LF, Shen YW, Lin NC, Shih YH, Shieh TM. Mechanistic insights into temoporfin-based photodynamic therapy: Ferroptosis as a critical regulator under normoxic and hypoxic conditions in head and neck cancer. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 267:113165. [PMID: 40267720 DOI: 10.1016/j.jphotobiol.2025.113165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Temoporfin is a second-generation photosensitizer used in photodynamic therapy (PDT) for the clinical treatment of head and neck cancer. However, its role in inhibiting cancer cell viability under normoxic and hypoxic conditions remains unclear. The oral squamous cell carcinoma (OSCC) cell lines, SAS and OECM-1 were cultured under normoxic or hypoxic conditions to investigate temoporfin-based PDT-induced cell death and the underlying mechanisms. Cell viability was analyzed using the MTT assay. Intracellular reactive oxygen species (ROS) levels, cell apoptosis, intracellular ROS, iron levels, lipid peroxidation, and glutathione (GSH) levels were assessed by flow cytometry. The expression of proteins related to oxidative stress, apoptosis, autophagy, and ferroptosis was verified by western blotting. Results showed that increasing the temoporfin dose, absorption time, and illumination time was positively correlated with the inhibition of oral cancer cells. Hypoxic conditions attenuated the toxicity of temoporfin in cancer cells. OECM-1 cells were more sensitive to temoporfin than SAS cells. Temoporfin-based PDT-induced ROS exhibited similar trends to oxidative stress-inducing enzymes under both normoxic and hypoxic conditions and triggered cell autophagy and ferroptosis. Administration of the ferroptosis inhibitor BRD4770 under normoxic conditions reversed temoporfin-based PDT-induced reductions in glutathione peroxidase 4 (GPx4), increasing in light chain 3-II (LC3-II) and cleaved poly (ADP-ribose) polymerase (cleaved-PARP). This study confirms that hypoxia weakens the anticancer effects of temoporfin-based PDT and that ferroptosis plays a key role in temoporfin-based PDT-mediated cancer cell inhibition.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Biobank, Chang Gung Memorial Hospital at Linkou, No.5, Fuxing Street, Guishan District, Taoyuan 33305, Taiwan; Graduate Institute of Health Industry Technology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No. 261, Wenhua 1st Rd., Guishan District, Taoyuan 33303, Taiwan; Liver Research Center, Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital at Linkou, No.5, Fuxing Street, Guishan District, Taoyuan 33305, Taiwan; Graduate Institute of Natural Products, Chang Gung University, No.259, Wenhua 1st Rd., Guishan District, Taoyuan 33302, Taiwan.
| | - Li-Fang Chou
- Kidney Research Center, Chang Gung Memorial Hospital, Tao-Yuan 33305, Taiwan
| | - Yen-Wen Shen
- School of Dentistry, China Medical University, No. 100, Section 1, Jingmao Road, Beitun District, Taichung 406040, Taiwan.
| | - Nan-Chin Lin
- Department of Oral and Maxillofacial Surgery, Show Chwan Memorial Hospital, No.542, Sec 1 Chung-shan Rd., Changhua 500. Taiwan
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, No.500, Lioufeng Rd., Wufeng, Taichung 413305, Taiwan.
| | - Tzong-Ming Shieh
- School of Dentistry, China Medical University, No. 100, Section 1, Jingmao Road, Beitun District, Taichung 406040, Taiwan; Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou District, Taipei 112304, Taiwan.
| |
Collapse
|
8
|
Du K, Hu W, Gao S, Gan J, You C, Zhang S. Identification of multiomics and immune infiltration-associated biomarkers for early gastric cancer: a machine learning-based diagnostic model development study. BMC Cancer 2025; 25:972. [PMID: 40450287 DOI: 10.1186/s12885-025-14396-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025] Open
Affiliation(s)
- Kewei Du
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Wenfei Hu
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Shan Gao
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Jianxin Gan
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Chongge You
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China.
| | - Shangdi Zhang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China.
| |
Collapse
|
9
|
Li Z, Lu Y, Wang L, Shi L, Wang T. Reactive oxygen species-dependent nanomedicine therapeutic modalities for gastric cancer. NANOSCALE ADVANCES 2025; 7:3210-3227. [PMID: 40308560 PMCID: PMC12038724 DOI: 10.1039/d5na00321k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025]
Abstract
Reactive oxygen species (ROS) play a double-edged role in gastric cancer (GC). Higher levels of ROS in tumor cells compared to normal cells facilitate tumor progression. Once ROS concentrations rise rapidly to toxic levels, they cause GC cell death, which is instead beneficial for GC treatment. Based on these functions, nano-delivery systems taking the therapeutic advantages of ROS have been widely employed in tumor therapy in recent years, overcoming the drawbacks of conventional drug delivery techniques, such as non-specific systemic effects. In this review, the precise impacts of ROS on GC have been detailed, along with ROS-based nanomedicine therapeutic schemes. These strategies mainly focused on the use of excess ROS in the tumor microenvironment for controlled drug release and a substantial enhancement of ROS concentrations for tumor killing. The challenges and opportunities for the advancement of these anticancer therapies are also emphasized.
Collapse
Affiliation(s)
- Zhiyan Li
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008 China
| | - Yanjun Lu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008 China
| | - Lulu Wang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008 China
| | - Liuyi Shi
- Yangzhou University Medical College Yangzhou 225001 China
| | - Tao Wang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008 China
| |
Collapse
|
10
|
Laino A, Gabellone C, Gómez Lobato M, Arrighetti F, Tau J, Rodríguez Gil S, Garcia CF. First study on the effect of UV radiation on spiders: biochemical analysis. JOURNAL OF INSECT PHYSIOLOGY 2025; 163:104821. [PMID: 40414342 DOI: 10.1016/j.jinsphys.2025.104821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 05/07/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Ultraviolet radiation is one of the environmental factors that impacts all living beings and can cause various types of damage. In this study, the effects of UV-B and UV-C radiation on the antioxidant response, reactive oxygen species levels, and potential histological and chromosomal damage were evaluated for the first time in the spider Misumenops maculissparsus at three developmental stages (juveniles J3, J4, and adults). Juveniles exhibited significant inhibition of the antioxidant enzymes superoxide dismutase, catalase, and glutathione-S-transferase under UV radiation exposure. In adults, no enzymatic activity changes were observed, but there were increases in ROS levels, particularly under UV-C exposure, and alterations in the cellular population analyzed by flow cytometry. Histological analysis revealed substantial pigment accumulation beneath the cuticle in juveniles and, to a lesser extent, in adults, which could serve as a natural barrier and antioxidant defense against UV radiation. Cytogenetic analysis determined for the first time in this species a diploid chromosome number of 2n = 23 and identified chromatin granules in UV radiation-exposed adults, suggesting UV radiation-induced chromatin damage. These multidisciplinary findings enhance our understanding of the physiological defenses that spiders employ when exposed to UV radiation at different developmental stages.
Collapse
Affiliation(s)
- A Laino
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner," (INIBIOLP), La Plata, Argentina
| | - C Gabellone
- Centro de Estudios Parasitológicos y Vectores (CEPAVE), La Plata, Argentina
| | - M Gómez Lobato
- Instituto de Fisiología Vegetal (INFIVE), La Plata, Argentina
| | - F Arrighetti
- CONICET-Museo Argentino de Ciencias Naturales Bernardino Rivadavia, Ciudad Autónoma de Buenos Aires, Argentina
| | - J Tau
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner," (INIBIOLP), La Plata, Argentina
| | - S Rodríguez Gil
- Centro de Estudios Parasitológicos y Vectores (CEPAVE), La Plata, Argentina
| | - C F Garcia
- Instituto de Investigaciones Bioquímicas de La Plata "Prof. Dr. Rodolfo R. Brenner," (INIBIOLP), La Plata, Argentina.
| |
Collapse
|
11
|
Liu X, Luo Q, Zhao Y, Ren P, Jin Y, Zhou J. The Ferroptosis-Mitochondrial Axis in Depression: Unraveling the Feedforward Loop of Oxidative Stress, Metabolic Homeostasis Dysregulation, and Neuroinflammation. Antioxidants (Basel) 2025; 14:613. [PMID: 40427494 PMCID: PMC12108521 DOI: 10.3390/antiox14050613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Emerging evidence links ferroptosis-mitochondrial dysregulation to depression pathogenesis through an oxidative stress-energy deficit-neuroinflammation cycle driven by iron overload. This study demonstrates that iron accumulation initiates ferroptosis via Fenton reaction-mediated lipid peroxidation, compromising neuronal membrane integrity and disabling the GPx4 antioxidant system. Concurrent mitochondrial complex I/IV dysfunction impairs ATP synthesis, creating an AMPK/mTOR signaling imbalance and calcium dyshomeostasis that synergistically impair synaptic plasticity. Bidirectional crosstalk emerges: lipid peroxidation derivatives oxidize mitochondrial cardiolipin, while mitochondrial ROS overproduction activates ACSL4 to amplify ferroptotic susceptibility, forming a self-reinforcing neurodegenerative loop. Prefrontal-hippocampal metabolomics reveal paradoxical metabolic reprogramming with glycolytic compensation suppressing mitochondrial biogenesis (via PGC-1α/TFAM downregulation), trapping neurons in bioenergetic crisis. Clinical data further show that microglial M1 polarization through cGAS-STING activation sustains neuroinflammation via IL-6/TNF-α release. We propose a "ferroptosis-mitochondrial fragmentation-metabolic maladaptation" triad as mechanistic subtyping criteria for depression. Preclinical validation shows that combinatorial therapy (iron chelators + SIRT3 agonists) rescues neuronal viability by restoring mitochondrial integrity and energy flux. This work shifts therapeutic paradigms from monoaminergic targets toward multimodal strategies addressing iron homeostasis, organelle dynamics, and metabolic vulnerability-a framework with significant implications for developing neuroprotective antidepressants.
Collapse
Affiliation(s)
- Xu Liu
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Qiang Luo
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Yulong Zhao
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Peng Ren
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Yu Jin
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Junjie Zhou
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou 341000, China
| |
Collapse
|
12
|
Gwozdzinski K, Pieniazek A, Gwozdzinski L. Nitroxides: Chemistry, Antioxidant Properties, and Biomedical Applications. Molecules 2025; 30:2159. [PMID: 40430331 PMCID: PMC12114102 DOI: 10.3390/molecules30102159] [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: 05/08/2025] [Accepted: 05/11/2025] [Indexed: 05/29/2025] Open
Abstract
Nitroxides are stable organic free radicals with a wide range of applications. They have found applications in chemistry, biochemistry, biophysics, molecular biology, and biomedicine as EPR/NMR imaging techniques. As spin labels and probes, they are used in electron paramagnetic resonance (EPR) spectroscopy in the study of proteins, lipids, nucleic acids, and enzymes, as well as for measuring oxygen concentration in cells and cellular organelles, as well as tissues and intracellular pH. Their unique redox properties have allowed them to be used as exogenous antioxidants. In this review, we have discussed the chemical properties of nitroxides and their antioxidant properties. Furthermore, we have considered their use as radioprotectors and protective agents in ischemia/reperfusion in vivo and in vitro. We also presented other applications of nitroxides in protecting cells and tissues from oxidative stress and in protein studies and discussed their use in EPR/MRI.
Collapse
Affiliation(s)
- Krzysztof Gwozdzinski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (K.G.); (A.P.)
| | - Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (K.G.); (A.P.)
| | - Lukasz Gwozdzinski
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-151 Lodz, Poland
| |
Collapse
|
13
|
Mohamad NA, Rahman AA, Kadir SHSA, Naes SM, Mazlan M, Makpol S. Hydroxychavicol in Combination with 5-Fluorouracil Induced Apoptosis by Inhibiting Purine Metabolism in HT-29 and DLD-1 Cell Lines. Chin J Integr Med 2025:10.1007/s11655-025-4133-1. [PMID: 40358878 DOI: 10.1007/s11655-025-4133-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/25/2024] [Indexed: 05/15/2025]
Abstract
OBJECTIVE To elucidate the effect of hydroxychavicol (HC) in combination with 5-fluorouracil (5-FU) on purine metabolism and apoptosis in colorectal cancer cell lines HT-29 and DLD-1. METHODS The viability of HT-29 and DLD-1 cells when treated with HC, (0-1,000 µmol/L) 5-FU (0-100 µmol/L) alone, and HC+5-FU for 24 and 48 h was determined. Hypoxanthine (HPX) and xanthine oxidoreductase (XOR) were evaluated, as well as reactive oxygen species (ROS) levels and mitochondrial membrane potential (MMP). The expression levels of genes including nucleoside transporters equilibrative nucleotide transport 1 and 2 (ENT1 and ENT2), the proapoptotic gene Caspase-3 (CASP3), and the anti-apoptotic gene BCL2 were analysed by quantitative polymerase chain reaction. RESULTS Both HPX and XOR levels in cells treated with HC+5-FU were significantly decreased (P<0.05) after 24 and 48 h compared to control cells. ROS levels in HT-29 and DLD-1 treated with HC+5-FU for 24 and 48 h were 26.2% and 21.4%, and 9.1% and 20.5%, respectively, significantly lower than control cells. MMP assays indicated mitochondrial depolarisation. In HT-29 cells, ENT1 and BCL2 were downregulated at 24 h, and CASP3 was upregulated at 48 h. In DLD-1 cells, ENT1 and ENT2 were downregulated, while CASP3 showed a transient decrease at 24 h. CONCLUSIONS The combination of HC + 5-FU demonstrated synergistic effects in HT-29 and DLD-1 cells, disrupting oxidative balance and purine metabolism, as reflected in reduced hypoxanthine levels, XOR activity, and ROS production. This treatment also induced mitochondrial membrane depolarisation and altered apoptosis-related gene expression, supporting its role in apoptosis induction.
Collapse
Affiliation(s)
- Noor Azleen Mohamad
- Department of Biochemistry & Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, 47000, Selangor, Malaysia
- Institute of Medical & Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, 47000, Selangor, Malaysia
| | - Amirah Abdul Rahman
- Department of Biochemistry & Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, 47000, Selangor, Malaysia.
| | - Siti Hamimah Sheikh Abdul Kadir
- Department of Biochemistry & Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, 47000, Selangor, Malaysia
| | - Safaa M Naes
- Department of Biochemistry & Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, 47000, Selangor, Malaysia
- Institute of Medical & Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, 47000, Selangor, Malaysia
- Faculty of Health and Life Sciences, Management and Science University, Shah Alam, 40100, Malaysia
| | - Musalmah Mazlan
- Department of Biochemistry & Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, 47000, Selangor, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kampus Kuala Lumpur, Cheras, Kuala Lumpur, 56000, Malaysia
| |
Collapse
|
14
|
Xi X, Qi Y, Zhang M, Yang P, Huang X. Unveiling 8,12-Dimethoxysanguinarine: A Potent Inhibitor of Breast Cancer Metastasis via Fibronectin 1 Downregulation. Chem Biodivers 2025; 22:e202402489. [PMID: 39676589 DOI: 10.1002/cbdv.202402489] [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: 10/01/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/17/2024]
Abstract
This study investigated the effects of 8,12-dimethoxysanguinarine (DSG) from Eomecon chionantha Hance on the malignant biological activity of breast cancer cells. RNA-sequencing measure analysis revealed that metastasis-related genes were significantly downregulated in DSG-treated MCF-7 cells. DSG significantly inhibits the migration ability in MCF-7 cells. Molecular docking studies demonstrated significant interactions between DSG and the Fibronectin 1 (FN1) protein, with a binding energy of -8.91 kcal/mol. Additionally, FN1 messenger RNA expression was significantly upregulated in 1085 breast tumor samples compared to normal tissue in the Cancer Genome Atlas Breast Invasive Carcinoma Collection dataset. DSG also suppressed MCF-7 cell metastasis by downregulating FN1 expression. Furthermore, DSG was identified as a promising candidate based on absorption distribution metabolism excretion toxicity and drug-likeness assessments. Combination studies indicated that DSG synergized with the conventional chemotherapeutic agent doxorubicin to suppress MCF-7 cell migration, as confirmed by wound-healing and transwell assays. Collectively, these findings suggest that DSG may serve as a potential agent for inhibiting breast cancer cell metastasis by decreasing FN1 expression.
Collapse
Affiliation(s)
- Xiuli Xi
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Yuxin Qi
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Mingli Zhang
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Peng Yang
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Xueshuang Huang
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
15
|
Liu K, Hu S, Wufuer R, Zhang Q, Qiu L, Zhang Z, Wang M, Zhang Y. Deficiency of DDI2 suppresses liver cancer progression by worsening cell survival conditions. Free Radic Biol Med 2025; 232:200-213. [PMID: 40049338 DOI: 10.1016/j.freeradbiomed.2025.03.003] [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: 12/22/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
The levels of reactive oxygen species (ROS) and the extent of ensuing DNA damage significantly influence cancer initiation and progression. Of crucial importance, the aspartate protease DDI2 has been proposed to play a pivotal role in monitoring intracellular ROS levels (to trigger oxidative eustress or distress), as well as in the oxidative DNA damage repair, through redox homeostasis-determining factor Nrf1 (encoded by NFE2L1). However, the specific role of DDI2 in the multi-step process resulting in the development and progression of liver cancer remains elusive to date. In the present study, we employed the CRISPR/Cas9 gene editing system to create two nuanced lines of DDI2 knockout (i.e., DDI2-/- and DDI2insG/-) from liver cancer cells. Subsequent experiments indicate that the knockout of DDI2 leads to increased ROS levels in hepatoma cells by downregulating two major antioxidant transcription factors Nrf1 and Nrf2 (encoded by NFE2L2), exacerbating endogenous DNA damages caused by ROS and not-yet-identified factors, thereby inhibiting cell proliferation and promoting apoptosis, and ultimately hindering in vivo malignant growth of xenograft tumor cells. Conversely, the restoration of DDI2 expression reverses the accumulation of ROS and associated DNA damage caused by DDI2 knockout, eliminating the subsequent inhibitory effects of DDI2 deficiency on both in vitro and in vivo growth of liver cancer cells. Collectively, these findings demonstrate that DDI2 deficiency impedes liver tumor growth by disrupting its survival environment, suggesting that DDI2 may serve as a novel therapeutic target for anti-cancer strategies aimed at modulating ROS or DNA damage processes.
Collapse
Affiliation(s)
- Keli Liu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Shaofan Hu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Jinfeng Laboratory, No. 313 Jinyue Road, Chongqing High-tech District, 401329, China; Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba, Chongqing, 400038, China
| | - Reziyamu Wufuer
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; School of Parmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Xinjiang, 830017, China
| | - Qun Zhang
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Lu Qiu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Zhengwen Zhang
- Laboratory of Neuroscience, Institute of Cognitive Neuroscience and School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, England, United Kingdom
| | - Meng Wang
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Jinfeng Laboratory, No. 313 Jinyue Road, Chongqing High-tech District, 401329, China; Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba, Chongqing, 400038, China.
| | - Yiguo Zhang
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; School of Life and Health Sciences, Fuyao University of Science and Technology, No. 104 Wisdom Avenue, Nanyu Town, Minhou County High-Tech District, Fuzhou, 350109, Fujian, China.
| |
Collapse
|
16
|
Park C, Cha HJ, Hong SH, Noh JS, Hong SH, Kim GY, Shim JH, Hyun JW, Choi YH. Paeoniflorin Protects Retinal Pigment Epithelial Cells from High Glucose-Induced Oxidative Damage by Activating Nrf2-Mediated HO-1 Signaling. Biomol Ther (Seoul) 2025; 33:518-528. [PMID: 40241343 PMCID: PMC12059356 DOI: 10.4062/biomolther.2025.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 04/18/2025] Open
Abstract
Oxidative stress due to hyperglycemia damages the functions of retinal pigment epithelial (RPE) cells and is a major risk factor for diabetic retinopathy (DR). Paeoniflorin is a monoterpenoid glycoside found in the roots of Paeonia lactiflora Pall and has been reported to have a variety of health benefits. However, the mechanisms underlying its therapeutic effects on high glucose (HG)-induced oxidative damage in RPE cells are not fully understood. In this study, we investigated the protective effect of paeoniflorin against HG-induced oxidative damage in cultured human RPE ARPE-19 cells, an in vitro model of hyperglycemia. Pretreatment with paeoniflorin markedly reduced HG-induced cytotoxicity and DNA damage. Paeoniflorin inhibited HG-induced apoptosis by suppressing activation of the caspase cascade, and this suppression was associated with the blockade of cytochrome c release to cytoplasm by maintaining mitochondrial membrane stability. In addition, paeoniflorin suppressed the HG-induced production of reactive oxygen species (ROS), increased the phosphorylation of nuclear factor erythroid 2-related factor 2 (Nrf2), a key redox regulator, and the expression of its downstream factor heme oxygenase-1 (HO-1). On the other hand, zinc protoporphyrin (ZnPP), an inhibitor of HO-1, abolished the protective effect of paeoniflorin against ROS production in HG-treated cells. Furthermore, ZnPP reversed the protective effects of paeoniflorin against HG-induced cellular damage and induced mitochondrial damage, DNA injury, and apoptosis in paeoniflorin-treated cells. These results suggest that paeoniflorin protects RPE cells from HG-mediated oxidative stress-induced cytotoxicity by activating Nrf2/HO-1 signaling and highlight the potential therapeutic use of paeoniflorin to improve the symptoms of DR.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-eui University, Busan 47340, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49104, Republic of Korea
| | - Su Hyun Hong
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Jeong Sook Noh
- Department of Food Science & Nutrition, Tongmyong University, Busan 48520, Republic of Korea
| | - Sang Hoon Hong
- Department of Internal Medicine, Dong-eui University College of Korean Medicine, Busan 614-052, Republic of Korea
| | - Gi Young Kim
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Jung-Hyun Shim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Yung Hyun Choi
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| |
Collapse
|
17
|
Attique I, Haider Z, Khan M, Hassan S, Soliman MM, Ibrahim WN, Anjum S. Reactive Oxygen Species: From Tumorigenesis to Therapeutic Strategies in Cancer. Cancer Med 2025; 14:e70947. [PMID: 40377005 DOI: 10.1002/cam4.70947] [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: 11/26/2024] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Reactive oxygen species (ROS), a class of highly reactive molecules, are closely linked to the pathogenesis of various cancers. While ROS primarily originate from normal cellular processes, external stimuli can also contribute to their production. Cancer cells typically exhibit elevated ROS levels due to disrupted redox homeostasis, characterized by an imbalance between antioxidant and oxidant species. ROS play a dual role in cancer biology: at moderate levels, they facilitate tumor progression by regulating oncogenes and tumor suppressor genes, inducing mutations, promoting proliferation, extracellular matrix remodeling, invasion, immune modulation, and angiogenesis. However, excessive ROS levels can cause cellular damage and initiate apoptosis, necroptosis, or ferroptosis. METHODS This review explores molecular targets involved in redox homeostasis dysregulation and examines the impact of ROS on the tumor microenvironment (TME). Literature from recent in vitro and in vivo studies was analyzed to assess how ROS modulation contributes to cancer development and therapy. RESULTS Findings indicate that ROS influence cancer progression through various pathways and cellular mechanisms. Targeting ROS synthesis or enhancing ROS accumulation in tumor cells has shown promising anticancer effects. These therapeutic strategies exhibit significant potential to impair tumor growth while also interacting with elements of the TME. CONCLUSION The ROS serve as both promoters and suppressors of cancer depending on their intracellular concentration. Their complex role offers valuable opportunities for targeted cancer therapies. While challenges remain in precisely modulating ROS for therapeutic benefit, they hold promise as synergistic agents alongside conventional treatments, opening new avenues in cancer management.
Collapse
Affiliation(s)
- Iqra Attique
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| | - Zahra Haider
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| | - Maha Khan
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Samina Hassan
- Department of Botany, Kinnaird College for Women University, Lahore, Pakistan
| | - Mohamed Mohamed Soliman
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, Taif, Saudi Arabia
- Biochemistry Department, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Sumaira Anjum
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| |
Collapse
|
18
|
Gao J, Zhang C, Hu LJ, Lin K, Zhou M, Zhu SX, Sun BL, Liu W, Shu XJ, Gan GP, Ye XS, Yang DS. Isolation and Characterization of Sesquiterpene Lactones From Syneilesis aconitifolia and Evaluation of Their Anti-Breast Cancer Activity. Chem Biodivers 2025; 22:e202402891. [PMID: 39641744 DOI: 10.1002/cbdv.202402891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/07/2024]
Abstract
Two undescribed sesquiterpene lactones (1 and 2), along with two known analogs (3 and 4), were isolated from Syneilesis aconitifolia. Their structures and absolute configurations were elucidated using spectroscopic analysis, ECD calculations, and single crystal x-ray diffraction. Compound 3 exhibited the highest cytotoxic activity against 4T1 cells, with an IC50 value of 10.89 µM. In addition, compound 3 significantly induced the apoptosis of 4T1 cells at the concentration of 20 µM. Further anticancer study showed that compound 3 distinctly increased the production of intracellular reactive oxygen species.
Collapse
Affiliation(s)
- Jie Gao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Cong Zhang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Li-Juan Hu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Kuan Lin
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Mei Zhou
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Shu-Xiu Zhu
- Department of Traditional Chinese Medicine, School of Medicine, Jianghan University, Wuhan, China
| | - Bin-Lian Sun
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Wei Liu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Xi-Ji Shu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Guo-Ping Gan
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Chinese Materia Medica Processing Engineering Center of Hubei Province, Hubei University of Chinese Medicine, Wuhan, China
| | - Xian-Sheng Ye
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - De-Sen Yang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Chinese Materia Medica Processing Engineering Center of Hubei Province, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Donghu New Technology Development Zone, Wuhan, China
| |
Collapse
|
19
|
Gao L, Lv G, Liu Z, Tian Y, Han F, Li L, Wang G, Zhang Y. Alcohol-induced C/EBP β-driven VIRMA decreases oxidative stress and promotes pancreatic ductal adenocarcinoma growth and metastasis via the m6A/YTHDF2/SLC43A2 pathway. Oncogene 2025; 44:1118-1132. [PMID: 39900725 DOI: 10.1038/s41388-025-03283-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 01/06/2025] [Accepted: 01/22/2025] [Indexed: 02/05/2025]
Abstract
N6-methyladenosine (m6A) plays a role in the development of tumors. However, the specific role of VIRMA, an RNA methyltransferase, in pancreatic ductal adenocarcinoma (PDAC) remains unclear. This study shows that VIRMA expression is elevated in PDAC. Increased VIRMA levels promoted PDAC growth and spread, while reducing VIRMA expression slowed these processes. VIRMA facilitated SLC43A2 mRNA degradation through an m6A-YTHDF2 pathway. The resulting decrease in SLC43A2 reduced phenylalanine absorption and oxidative stress, further driving PDAC progression. Furthermore, alcohol increased C/EBP β expression, which bound to VIRMA's promoter, enhancing its transcription. These findings suggest a connection between alcohol consumption, m6A modifications, and phenylalanine absorption in PDAC progression, offering a new approach to combat this disease.
Collapse
Affiliation(s)
- Lei Gao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gaoyuan Lv
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ziying Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yitong Tian
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Fang Han
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Yuhua Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
20
|
Tang S, Li J, Tian W, Feng Y, Deng Y, Tan Z, Han Z, He H, Wu Y, Huang C, Ning K, Liu F, Luo H, Cai S, Ye J, Zhong W. Characterization of the Biochemical Recurrence Prediction Ability and Progression Correlation of Peroxiredoxins Family in Prostate Cancer Based on Integrating Single-Cell RNA-Seq and Bulk RNA-Seq Cohorts. Cancer Med 2025; 14:e70855. [PMID: 40281661 PMCID: PMC12031674 DOI: 10.1002/cam4.70855] [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: 10/11/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION The peroxiredoxins (PRDXs) family plays a crucial role in balancing reactive oxygen species (ROS) levels in tumor cells. However, its potential role in prognosis and therapy response of prostate cancer (PCa) remains unknown. METHODS In this study, we utilized 2 public single-cell RNA datasets and 8 bulk-RNA datasets to investigate the clinical value of six PRDXs family members in PCa. Expression comparison, biochemical recurrence analysis, and therapy response analysis were measured. Pathway enrichments were utilized to predict the potential down-stream pathway it may involve. In vitro experiments were used to validate the function of PRDX5 in the progression of castration-resistant prostate cancer (CRPC) cell lines. RESULT Among the PRDXs family, PRDX5 was most related to the advancement of prostate cancer. A nomogram integrating the expression of PRDX5 with clinical features was developed to better predict clinical outcomes in PCa patients compared to 30 published signatures. Immunohistochemistry was used to verify that PRDX5 expression was higher in advanced levels of PCa tissue. Gene Set Enrichment Analysis (GSEA) and pathway predictive analysis revealed that the PRDX5 related genes were mainly relevant to ROS Pathway, Mitochondria-related functions, cellular respiration, and oxidative phosphorylation. In vitro cell proliferation assays, ROS determination assay, and apoptosis assay together revealed that depletion of PRDX5 induces apoptosis via ROS accumulation in CRPC cells. Moreover, the expression of PRDX5 in CRPC cells also affects the sensitivity to the ARSI therapy. CONCLUSION This study offers new evidence for determining that the expression of PRDX5 is associated with advanced tumor grade, poor prognosis, and suboptimal response to multiple therapies in PCa within the PRDXs family. Last but not least, our study provides new insights into precision medicine in PCa and provides a reference for further research on PRDX5.
Collapse
Affiliation(s)
- Shan Tang
- Urology DepartmentThe Central Hospital of ShaoyangShaoyangChina
| | - Jinchuang Li
- Department of UrologyGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
- Guangdong Key Laboratory of Clinical Molecular Medicine and DiagnosticsGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Weicheng Tian
- Guangdong Provincial Key Laboratory of UrologyThe First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical UniversityGuangzhouChina
| | - Yuanfa Feng
- Guangdong Provincial Key Laboratory of UrologyThe First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical UniversityGuangzhouChina
- Guangzhou National LaboratoryGuangzhouChina
| | - Yulin Deng
- Department of UrologyThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanChina
| | - Zeheng Tan
- Department of UrologyGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
- Guangdong Key Laboratory of Clinical Molecular Medicine and DiagnosticsGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Zhaodong Han
- Department of UrologyGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
- Guangdong Key Laboratory of Clinical Molecular Medicine and DiagnosticsGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Huichan He
- Guangdong Provincial Key Laboratory of UrologyThe First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical UniversityGuangzhouChina
| | - Yongding Wu
- Guangdong Key Laboratory of Clinical Molecular Medicine and DiagnosticsGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Chuyang Huang
- Urology DepartmentThe Central Hospital of ShaoyangShaoyangChina
| | - Keping Ning
- Urology DepartmentThe Central Hospital of ShaoyangShaoyangChina
| | - Feng Liu
- Urology DepartmentThe Central Hospital of ShaoyangShaoyangChina
| | - Hongwei Luo
- Urology DepartmentThe Central Hospital of ShaoyangShaoyangChina
| | - Shanghua Cai
- Guangdong Provincial Key Laboratory of UrologyThe First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical UniversityGuangzhouChina
- Guangzhou National LaboratoryGuangzhouChina
| | - Jianheng Ye
- Department of UrologyGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
- Guangdong Key Laboratory of Clinical Molecular Medicine and DiagnosticsGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Weide Zhong
- Department of UrologyGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
- Guangdong Key Laboratory of Clinical Molecular Medicine and DiagnosticsGuangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
- Guangdong Provincial Key Laboratory of UrologyThe First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical UniversityGuangzhouChina
- Guangzhou National LaboratoryGuangzhouChina
| |
Collapse
|
21
|
Alzahrani SM, Al Doghaither HA, Alkhatabi HA, Basabrain MA, Pushparaj PN. Propranolol and Capecitabine Synergy on Inducing Ferroptosis in Human Colorectal Cancer Cells: Potential Implications in Cancer Therapy. Cancers (Basel) 2025; 17:1470. [PMID: 40361395 PMCID: PMC12071015 DOI: 10.3390/cancers17091470] [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: 02/22/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Colorectal cancer (CRC) is a significant global health issue with rising incidence and mortality rates. In oncology, drug repurposing has emerged as a promising therapeutic strategy in conjunction with conventional treatments. This study aimed to evaluate the potential of repurposing propranolol (PRO), a beta blocker, for the treatment of CRC cell lines (HCT-116 and HT-29), both as a monotherapy and in combination with capecitabine (CAP). METHODS Effects of mono- and combination therapies on viability, combination index, morphology, and cell death induction of CRC cells were assessed. Transcriptome analysis of HT-29 cells was performed using RNA sequencing. Metabolite profiling was conducted, and changes in biochemical parameters were evaluated using flow cytometry and biochemical analyses. RESULTS The combination index showed that HT-29 cells were the most responsive to the combined treatment, even with PIK3CA, B-RAF (V600E), and TP53 mutations. Moreover, ferroptosis was synergistically activated in the combined group of HT-29 in comparison to control. Furthermore, we observed an increase in OXPHOS metabolites, along with elevated intracellular and mitochondrial ROS, disruption of mitochondrial membrane potential, and greater levels of malondialdehyde (MDA) in the HT-29 combined group, which are the features of ferroptosis. Furthermore, ferroptosis induction was coupled with necroptosis, as indicated by RNA-sequencing data. Combination therapy inhibited cell migration and enhanced the immune response of HT-29 cells. CONCLUSIONS These findings suggest that PRO is promising as a potential adjuvant therapy in combination with CAP for the treatment of CRC. Only HT-29 cells with the B-RAF (V600E) mutation showed promising findings in this study.
Collapse
Affiliation(s)
- Shiekhah Mohammad Alzahrani
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
- Institute of Genomic Medicine Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
| | | | - Hind Ali Alkhatabi
- Department of Biological Science, College of Science, University of Jeddah, Jeddah P.O. Box 21589, Saudi Arabia
| | - Mohammad Abdullah Basabrain
- Institute of Genomic Medicine Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
| | - Peter Natesan Pushparaj
- Institute of Genomic Medicine Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah P.O. Box 21589, Saudi Arabia
| |
Collapse
|
22
|
Yang Y, Wang H, Xue Q, Peng W, Zhou Q. New advances of natural products in non-small cell lung cancer: From mechanisms to therapies. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119636. [PMID: 40120701 DOI: 10.1016/j.jep.2025.119636] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE With the rise of immunotherapy, the treatment approach for non-small cell lung cancer (NSCLC) has undergone revolutionary changes. However, the prognosis for NSCLC patients has not been significantly improved due to the development of acquired drug resistance. Therefore, there is an urgent need to develop new and more effective drugs for treating NSCLC or improving tumor treatment resistance. Traditional Chinese medicine (TCM) has been gradually incorporated into the combined treatment of NSCLC. Its active components (also known as natural products) exhibit novel structures, multi-target effects, diverse pathways, minimal toxicity, and varied biological activities, which play a therapeutic role in various diseases. Thus, natural products hold great potential for future clinical applications. AIM OF THE STUDY Screening main traditional plants widely used in NSCLC and their derived natural products, as well as exploring the mechanisms by which these natural products act on NSCLC-particularly focusing on their applications-can provide valuable insights for the development of therapeutic drugs targeting NSCLC. METHODS A comprehensive, computerized literature search was conducted in PubMed, Embase, Web of Science, Cochrane Library, CNKI Scholar, the American Chemical Abstracts, and Wanfang Database up to June 2024, using the following keywords: "traditional Chinese medicine", "herbal medicine", "medicinal plants", and "herbal", paired with terms such as "non-small cell lung cancer", "therapy", "natural products", and "active ingredient". RESULTS Summarizing current research findings, we discovered eleven medicinal plants containing a total of fourteen natural products. Natural products have a significant impact on tumor progression in NSCLC, including apotosis, autophagy, pyrotosis, cell-cycle arrest and metasis. Moreover, natural products can modulate the activities of various immune cells and reshape the immune microenvironment. Combined with conventional cancer treatments, natural products demonstrate promising therapeutic effects and effectively reverse drug resistance. Furthermore,the use of nano-drug delivery systems to address limitations associated with natural products. CONCLUSIONS This review summarizes eleven medicinal plants containing a total of fourteen natural products that can enhance NSCLC treatment and indicates their action mechanisms. Furthermore, we also discuss limitations of natural products and explore the use of nano-drug delivery systems to address limitations associated with natural products.
Collapse
Affiliation(s)
- Yuening Yang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Haolei Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Qianqian Xue
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Wenbei Peng
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Qiong Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.
| |
Collapse
|
23
|
Chen Q, Feng G, Shen Y, Li X, Pei Q, Wang H, Tian L, Cao Y, Wu J, Yang H, Mu L. An Anionic Cathelicidin Exerts Antimelanoma Effects in Mice by Promoting Pyroptosis. J Med Chem 2025; 68:8618-8633. [PMID: 40207383 DOI: 10.1021/acs.jmedchem.5c00281] [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]
Abstract
While cationic antimicrobial peptides (AMPs) are extensively studied for antitumor effects, anionic AMPs remain underexplored. Notably, no amphibian-derived anionic cathelicidins with antitumor activity have been reported. This study identifies Boma-CATH, a novel anionic cathelicidin (net charge-3) from Bombina maxima skin, which suppresses melanoma growth in mice and triggers pyroptosis-like morphological changes in A375 cells via the NLRP3/Caspase-1/GSDMD pathway. Further investigation revealed that ROS played a crucial role in promoting pyroptosis, as NAC (ROS scavenger) and Ac-YVAD-cmk (Caspase-1 inhibitor) reversed cell death and reduced LDH/IL-1β release in vitro and in vivo. GSDMD knockdown further validated its role. Additionally, Boma-CATH inhibited A375 cell proliferation, migration, and invasion, demonstrating dual antitumor mechanisms: pyroptosis induction and metastasis suppression. Importantly, Boma-CATH caused no adverse effects in mice, highlighting its therapeutic safety. These findings position Boma-CATH as a promising melanoma treatment and expand the mechanistic understanding of anionic AMPs in oncology.
Collapse
Affiliation(s)
- Qian Chen
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Guizhu Feng
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yan Shen
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Xiang Li
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Qiqi Pei
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Hanying Wang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Li Tian
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yuanyuan Cao
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jing Wu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Hailong Yang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Lixian Mu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| |
Collapse
|
24
|
Bhattacharyya T, Das P, Ansari A, Mohan AA, Chandra Y, Narayan KP, Banerjee R. Glucocorticoid Receptor-Targeted Nanoliposome for STAT3 Inhibition-Led Myeloid-Derived Suppressor Cell Modulation and Efficient Colon Cancer Treatment. ACS APPLIED BIO MATERIALS 2025; 8:3185-3204. [PMID: 40162961 DOI: 10.1021/acsabm.5c00002] [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] [Indexed: 04/02/2025]
Abstract
STAT3 is an important protein responsible for cellular proliferation, motility, and immune tolerance and is hyperactive in colorectal cancer, instigating metastasis, cellular proliferation, migration, as well as inhibition. It helps in proliferation of myeloid-derived suppressor cells (MDSCs), which within the tumor microenvironment (TME) suppress T cells to encourage tumor growth, metastasis, and resistance to immunotherapy, besides playing dynamic role in regulating macrophages within the tumor. Thus, MDSC is a potential target to augment immune surveillance within the TME. Herein, we report targeting both colorectal cancer and MDSCs using a glucocorticoid receptor (GR)-targeted nanoliposomal formulation carrying GR-ligand, dexamethasone (Dex), and a STAT3 inhibitor, niclosamide (N). Our main objective was to selectively inhibit STAT3, the key immunomodulatory factor in most TME-associated cells including MDSCs, and also repurpose the use of this antihelminthic, low-cost drug N for cancer treatment. The resultant formulation D1XN exhibited better tumor regression and survivability compared to GR nontargeted formulation. Further, bone marrow cell-derived MDSCs were engineered by D1XN treatment ex vivo and were inoculated back to tumor-bearing mice. Significant tumor growth inhibition with enhanced antiproliferative immune cell signatures, such as T cell infiltration, decrease in Treg cells, and increased M1/M2 macrophage ratio within the TME were observed. This reveals the effectiveness of engineered MDSCs to modulate tumor surveillance besides reversing the aggressiveness of the tumor. Therefore, D1XN and D1XN-mediated engineered MDSCs alone or in combination can be considered as potent selective chemo-immunotherapeutic nanoliposomal agent(s) against colorectal cancer.
Collapse
Affiliation(s)
- Tithi Bhattacharyya
- Division of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pritam Das
- Division of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Aasia Ansari
- Division of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Adrij A Mohan
- Department of Biotechnology, Manipal Institute of Technology, Manipal, Karnataka 576104, India
| | - Yogesh Chandra
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, BITS Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad 500078, India
| | - Rajkumar Banerjee
- Division of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
25
|
Ramushu P, Mangoakoane DD, Makola RT, Matsebatlela TM. Lithium Induces Oxidative Stress, Apoptotic Cell Death, and G2/M Phase Cell Cycle Arrest in A549 Lung Cancer Cells. Molecules 2025; 30:1797. [PMID: 40333785 PMCID: PMC12029159 DOI: 10.3390/molecules30081797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
Lithium has been identified more than six decades ago as a preferred treatment option for manic depression. Due to its affordability, stability, minimal side effects, and immunomodulatory effects, recent studies on lithium have focused on its potential anticancer properties and possible mechanisms of action. Lung cancer ranks the highest as the main cause of death in males and has high mortality rates with low survival rates. In this study, lung adenocarcinoma (A549) cells were treated with various concentrations of lithium chloride to evaluate its inflammatory and anticancer properties. The in vitro cytotoxic effects of lithium chloride were assessed using the MTT [3-(4, 5-dimethythiazol-2-yl)-2, 5-diphenyltetrazolium bromide] assay, Muse® cell death, and cell cycle analysis. The nitric oxide and oxidative stress flow cytometry Muse® assays were used to monitor inflammation profiles of lithium-treated lung adenocarcinoma cells. The MTT viability assay showed the safe use of LiCl on the noncancerous RAW 264.7 macrophage cells below a concentration of 40 mM. Lithium reduced cell viability, induced late apoptotic cell death, and disrupted normal cell cycle progression in a dose-dependent manner, leading to cell cycle arrest in the S and G2/M phases of A549 cells. The induction of cell death by lithium in A549 cells is accompanied by increased ROS and nitric oxide production. This study shows that lithium chloride possesses some immunomodulatory cytotoxic effects on A549 lung cancer cells and can be further investigated for use in lung cancer treatment.
Collapse
Affiliation(s)
- Pearl Ramushu
- Department of Biochemistry, Microbiology and Biotechnology, Faculty of Science and Agriculture, University of Limpopo, P/bag x1106, Sovenga 0727, South Africa
| | | | | | | |
Collapse
|
26
|
Alrumaihi F, Babiker AY, Khan A. Lipid-Based Nanoformulations of [6]-Gingerol for the Chemoprevention of Benzo[a] Pyrene-Induced Lung Carcinogenesis: Preclinical Evidence. Pharmaceuticals (Basel) 2025; 18:574. [PMID: 40284009 PMCID: PMC12030401 DOI: 10.3390/ph18040574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: [6]-Gingerol ([6]-G), a bioactive compound derived from Zingiber officinale (ginger), exhibits strong anticancer potential but is hindered by poor aqueous solubility and low bioavailability. This study aimed to develop and evaluate PEGylated liposomal [6]-G (6-G-Lip) to enhance its stability, bioavailability, and chemopreventive efficacy in benzo[a]pyrene (BaP)-induced lung carcinogenesis. Methods: 6-G-Lip was synthesized using a modified thin-film hydration technique and characterized for size, polydispersity index (PDI), zeta potential, encapsulation efficiency (EE%), and release kinetics. The chemopreventive effects were assessed in BaP-induced lung cancer in Swiss albino mice, with prophylactic 6-G-Lip administration from two weeks before BaP exposure through 21 weeks. Cancer biomarkers, antioxidant enzyme activity, reactive oxygen species (ROS) generation, induction of apoptosis, and histopathological alterations were analyzed. Results: 6-G-Lip exhibited a particle size of 129.7 nm, a polydispersity index (PDI) of 0.16, a zeta potential of -18.2 mV, and an encapsulation efficiency (EE%) of 91%, ensuring stability and effective drug loading. The formulation exhibited a controlled release profile, with 26.5% and 47.5% of [6]-G released in PBS and serum, respectively, at 72 h. 6-G-Lip significantly lowered cancer biomarkers, restored antioxidant defenses (SOD: 5.60 U/min/mg protein; CAT: 166.66 μm H2O2/min/mg protein), reduced lipid peroxidation (MDA: 3.3 nm/min/mg protein), and induced apoptosis (42.2%), highlighting its chemopreventive efficacy. Conclusions: This study is the first to prepare, characterize, and evaluate PEGylated [6]-G-Lip for the chemoprevention of lung cancer. It modulates oxidative stress, restores biochemical homeostasis, and selectively induces apoptosis. These findings support 6-G-Lip as a promising nanotherapeutic strategy for cancer prevention.
Collapse
Affiliation(s)
- Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (F.A.); (A.Y.B.)
| | - Ali Yousif Babiker
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (F.A.); (A.Y.B.)
| | - Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
27
|
Yi F, Tao S, Wu H. Bilirubin metabolism in relation to cancer. Front Oncol 2025; 15:1570288. [PMID: 40291905 PMCID: PMC12021636 DOI: 10.3389/fonc.2025.1570288] [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: 02/03/2025] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
Bilirubin, a metabolite of hemoglobin, was long thought to be a harmful waste product, but recent studies have found it to have antioxidant and anti-tumor effects. With the extensive research on the mechanism of malignant tumor development, the antioxidant effect of bilirubin is increasingly becoming a hotspot in anti-cancer research. At present, there are two main views on the relationship between bilirubin and cancer, namely, its pro-cancer and anti-cancer effects, and in recent years, studies on the relationship between bilirubin and cancer have not been systematically summarized, which is not conducive to the further investigation of the role of bilirubin on cancer. To understand the multifaceted role of bilirubin in tumorigenesis as well as to develop more effective and affordable antitumor therapies, this review provides an overview of the effects of bilirubin on tumors in terms of oxidative, inflammatory, and cellular signaling pathways, as well as the resulting therapeutic ideas and approaches.
Collapse
Affiliation(s)
- Fengyun Yi
- Department of Traditional Chinese Medicine, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Siyu Tao
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongze Wu
- Department of Traditional Chinese Medicine, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
| |
Collapse
|
28
|
Yang Y, Zhu X, Sun D, Fan J. Lnc-CNNM3-DT as a protective factor in cervical cancer: regulation of LIAS expression and intracellular copper levels. Front Oncol 2025; 15:1571788. [PMID: 40265013 PMCID: PMC12011582 DOI: 10.3389/fonc.2025.1571788] [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: 02/06/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025] Open
Abstract
Background Cervical cancer (CC) is the fourth leading cause of cancer-related death in women globally.While early screening has reduced mortality, tumor metastasis remains a significant concern, particularly in developing countries. Recent studies have identified cuproptosis, a copper-dependent cell death mechanism, as a potential factor in tumor progression. Long non-coding RNAs (lncRNAs) are key regulators of tumor progression. This study investigates the role of cuproptosis-related lncRNA (CRL) CNNM3-DT in CC, focusing on its impact on LIAS expression, intracellular copper levels, and tumor progression. Methods We analyzed the expression of lnc-CNNM3-DT and LIAS in clinical samples and CC cell lines using Real-time Polymerase Chain Reaction (RT-qPCR), Western blot, and immunohistochemistry (IHC). Functional assays, including CCK-8, wound healing, transwell invasion, and flow cytometry, were used to evaluate the effects of lnc-CNNM3-DT overexpression on cell proliferation, migration, invasion, and apoptosis. Intracellular copper ion levels were measured, and correlations between lnc-CNNM3-DT, LIAS, and clinicopathological features were analyzed. Results Lnc-CNNM3-DT expression was significantly higher in paracancerous tissues and normal cervical epithelial cells compared to tumor tissues and CC cell lines. Overexpression of lnc-CNNM3-DT suppressed proliferation, migration, and invasion of HeLa and SiHa cells while enhancing apoptosis. Additionally, lnc-CNNM3-DT overexpression downregulated LIAS expression and decreased intracellular copper ion levels. Correlation analysis indicated that lnc-CNNM3-DT expression was negatively associated with tumor diameter and depth of invasion, while LIAS expression showed no significant correlation with clinicopathological features. Conclusion Our findings suggest that lnc-CNNM3-DT functions as a protective factor in CC by inhibiting tumor progression through downregulation of LIAS expression and reduction of intracellular copper levels. These results highlight lnc-CNNM3-DT as a potential biomarker and therapeutic target in CC.
Collapse
Affiliation(s)
- Ying Yang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Gynecology, Yulin First People’s Hospital (The Sixth Affiliated Hospital of Guangxi Medical University), Yulin, Guangxi, China
| | - Xuehong Zhu
- Department of Reproductive Medicine, Reproductive Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Dan Sun
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiangtao Fan
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
29
|
Dai X, Xi M, Li J. Cancer metastasis: molecular mechanisms and therapeutic interventions. MOLECULAR BIOMEDICINE 2025; 6:20. [PMID: 40192949 PMCID: PMC11977077 DOI: 10.1186/s43556-025-00261-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: 09/13/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
The metastatic cascade is a complicated process where cancer cells travel across multiple organs distant from their primary site of onset. Despite the wide acceptance of the 'seed and soil' theory, mechanisms driving metastasis organotropism remain mystery. Using breast cancer of different subtypes as the disease model, we characterized the 'metastatic profile of cancer cells' and the 'redox status of the organ microenvironment' as the primary determinants of cancer metastasis organotropism. Mechanically, we identified a positive correlation between cancer metabolic plasticity and stemness, and proposed oxidative stress as the selection power of cancer cells succeeding the metastasis cascade. Therapeutically, we proposed the use of pro-oxidative therapeutics in ablating cancer cells taking advantages of this fragile moment during metastasis. We comprehensively reviewed current pro-oxidative strategies for treating cancers that cover the first line chemo- and radio-therapies, approaches relying on naturally existing power including magnetic field, electric field, light and sound, nanoparticle-based anti-cancer composites obtained through artificial design, as well as cold atmospheric plasma as an innovative pro-oxidative multi-modal modality. We discussed possible combinations of pro-oxidative approaches with existing therapeutics in oncology prior to the forecast of future research directions. This paper identified the fundamental mechanics driving metastasis organotropism and proposed intervention strategies accordingly. Insights provided here may offer clues for the design of innovative solutions that may open a new paradigm for cancer treatment.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| | - Ming Xi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Jitian Li
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Henan Province, Zhengzhou, 450000, China
| |
Collapse
|
30
|
Chen Z, Niu K, Li M, Deng Y, Zhang J, Wei D, Wang J, Zhao Y. GCLC desuccinylation regulated by oxidative stress protects human cancer cells from ferroptosis. Cell Death Differ 2025:10.1038/s41418-025-01505-8. [PMID: 40188196 DOI: 10.1038/s41418-025-01505-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Tumor cells evolve strong antioxidant capacities to counteract the abnormal high level of reactive oxygen species (ROS) in the tumor microenvironment. Glutamate-cysteine ligase catalyzing subunit (GCLC) for synthesis of antioxidant glutathione (GSH) represents the key enzyme to maintain redox homeostasis of tumor cells, however, whether its activity is regulated by posttranslational modifications, such as succinylation, remains to be clarified. Here, we demonstrate the existence of succinylation modification on GCLC by in vitro and in vivo assays. NAD-dependent deacetylase Sirtuin-2 (SIRT2) serves as the desuccinylase and catalyzes GCLC desuccinylation at sites of K38, K126, and K326. Specifically, GCLC directly interacts with SIRT2, which can be substantially enhanced upon ROS treatment. This strengthened association results in GCLC desuccinylation and activation, consequently promoting GSH synthesis and rendering cancer cells resistant to ferroptosis induction. Depletion of SIRT2 decreases total GSH level and meanwhile increases the cellular susceptibility to ferroptosis, which can mostly be rescued by introducing wild-type GCLC, but not its 3K-E mutant. We further demonstrated that histone acetyltransferase P300 serves as the succinyltransferase of GCLC, and their association is remarkably decreased after ROS treatment. Thus, SIRT2-regulated GCLC succinylation represents an essential signaling axis for cancer cells to maintain their redox balance in coping with oxidative stress-induced ferroptosis.
Collapse
Affiliation(s)
- Zixiang Chen
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Kaifeng Niu
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Mengge Li
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuchun Deng
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ji Zhang
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Di Wei
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Jiaqi Wang
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongliang Zhao
- China National Center for Bioinformation, Beijing, China.
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
31
|
Hao MY, Li HJ, Han HS, Chu T, Wang YW, Si WR, Jiang QY, Wu DD. Recent advances in the role of gasotransmitters in necroptosis. Apoptosis 2025; 30:616-635. [PMID: 39833633 DOI: 10.1007/s10495-024-02057-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Necroptosis is a finely regulated programmed cell death process involving complex molecular mechanisms and signal transduction networks. Among them, receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like protein are the key molecules regulating this process. In recent years, gasotransmitters such as nitric oxide, carbon monoxide and hydrogen sulfide have been suggested to play a regulatory role in necroptosis. This paper reviews the evidence that these gasotransmitters are involved in the regulation of necroptosis by influencing the production of reactive oxygen species, regulating the modification of S subunits of RIPK1 and RIPK3, regulating inflammatory mediators, and signal transduction. In addition, this review explores the potential therapeutic applications of these gasotransmitters in pathological conditions such as cardiovascular disease and ischemia-reperfusion injury. Although some studies have revealed the important role of gasotransmitters in necroptosis, the specific mechanism of action is still not fully understood. Future research is needed to further elucidate the molecular mechanisms of gasotransmitters in precisely regulating necroptosis, which will help develop new therapeutic strategies to prevent and treat related diseases.
Collapse
Affiliation(s)
- Meng-Yuan Hao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Hong-Jie Li
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Hang-Shen Han
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ti Chu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yan-Wen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Wei-Rong Si
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
32
|
Xu M, Li W, Xu R, Liu L, Wu Z, Li W, Ma C, Xue L. Gp93 safeguards tissue homeostasis by preventing ROS-JNK-mediated apoptosis. Redox Biol 2025; 81:103537. [PMID: 39965405 PMCID: PMC11875814 DOI: 10.1016/j.redox.2025.103537] [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: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Reactive oxygen species (ROS) play a pivotal role in maintaining tissue homeostasis, yet their overabundance can impair normal cellular functions, induce cell death, and potentially lead to neurodegenerative disorders. This study identifies Drosophila Glycoprotein 93 (Gp93) as a crucial factor that safeguards tissue homeostasis and preserves normal neuronal functions by preventing ROS-induced, JNK-dependent apoptotic cell death. Firstly, loss of Gp93 induces JNK-dependent apoptosis primarily through the induction of ROS. Secondary, neuro-specific depletion of Gp93 results in ROS-JNK-mediated neurodegeneration. Thirdly, overexpression of Gp93 effectively curtails oxidative stress and neurodegeneration caused by paraquat exposure or the aging process. Furthermore, these functions of Gp93 can be substituted by its human ortholog, HSP90B1. Lastly, depletion of HSP90B1 in cultured human cells triggers ROS production, JNK activation, and apoptosis. Thus, this study not only unveils a novel physiological function of Gp93, but also provides valuable insights for understanding the physiological and pathological functions of human HSP90B1.
Collapse
Affiliation(s)
- Meng Xu
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Wanzhen Li
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Ruihong Xu
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Lixia Liu
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Zhihan Wu
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Wenzhe Li
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Chao Ma
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Lei Xue
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai 10th People's Hospital, 200072, Shanghai, China.
| |
Collapse
|
33
|
Xi D, Yang Y, Guo J, Wang M, Yan X, Li C. Single-cell sequencing and spatial transcriptomics reveal the evolution of glucose metabolism in hepatocellular carcinoma and identify G6PD as a potential therapeutic target. Front Oncol 2025; 15:1553722. [PMID: 40201344 PMCID: PMC11975570 DOI: 10.3389/fonc.2025.1553722] [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: 12/31/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Background Glucose metabolism reprogramming provides significant insights into the development and progression of malignant tumors. This study aims to explore the temporal-spatial evolution of the glucose metabolism in HCC using single-cell sequencing and spatial transcriptomics (ST), and validates G6PD as a potential therapeutic target for HCC. Methods We collected single-cell sequencing data from 7 HCC and adjacent non-cancerous tissues from the GSE149614 database, and ST data from 4 HCC tissues from the HRA000437 database. Pseudotime analysis was performed on the single-cell data, while ST data was used to analyze spatial metabolic activity. High-throughput sequencing and experiments, including wound healing, CCK-8, and transwell assays, were conducted to validate the role and regulatory mechanisms of G6PD in HCC. Results Our study identified a progressive upregulation of PPP-related genes during tumorigenesis. ST analysis revealed elevated PPP metabolic scores in the central and intermediate tumor regions compared to the peripheral zones. High-throughput sequencing and experimental validation further suggested that G6PD-mediated regulation of HCC cell proliferation, migration, and invasion is likely associated with glutathione metabolism and ROS production. Finally, Cox regression analysis cofirmed G6PD as an independent prognostic factor for overall survival in HCC patients. Conclusion Our study provides novel insights into the changes in glucose metabolism in HCC from both temporal and spatial perspectives. We experimentally demonstrated that G6PD regulates proliferation, migration, and invasion in HCC and propose G6PD as a prognostic marker and therapeutic metabolic target for the HCC.
Collapse
Affiliation(s)
- Deyang Xi
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yinshuang Yang
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiayi Guo
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mengjiao Wang
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xuebing Yan
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chunyang Li
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
34
|
Devangan P, Sharma A, Wadate N, Mourya A, Madan J. Eudragit S 100 Assisted Molecular Solid Dispersion of Andrographolide Tendered Augmented Drug Delivery and Apoptosis in Human Colon Cancer, HT-29 Cells. AAPS PharmSciTech 2025; 26:82. [PMID: 40069524 DOI: 10.1208/s12249-025-03073-z] [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] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
Colorectal cancer is the second most common cause of death due to growing incidence. Andrographolide (AGD) induces apoptosis in colorectal cancer cells; however, oral administration of AGD is associated with hindered aqueous solubility (3.29 ± 0.73-μg.mL-1) and bioavailability of 15.87 ± 3.84%. Therefore, in the current investigation, AGD was amalgamated with Eudragit S100 (EUS100) to engineer a molecular amorphous solid dispersion (EUSD). EUSD4, an optimized molecular solid dispersion showed ~ 5.90 and ~ 7.14-fold augmentations in solubility at pH ~ 6.8 and ~ 7.4, respectively as compared to AGD alone. The% assay and drug loading were respectively measured to be 96.01 ± 3.52% and 19.85 ± 0.65%. ATR and 1H-NMR spectroscopies confirmed that the -OH group of AGD formed an intermolecular hydrogen bond with the -C = O of EUS100. Moreover, a hallo pattern of PXRD, the disappearing of an endothermic peak in DSC, the absence of a birefringence pattern under polarized light, and disorders in the initial particle shape confirmed the amorphous state of EUSD4. In addition, a ~ 4.70- and ~ 2.94-fold enhancement in dissolution profile in simulated intestinal fluid (SIF, pH ~ 6.8) and simulated colonic fluid (SCF,pH ~ 7.4) of EUSD4 suggested amendment in the hydrophilicity, wettability properties, and dissolution rate. Furthermore, the IC50 of EUSD4 was ~ 1.42-fold higher than AGD, indicating improvement in anticancer efficacy against HT-29 cells. EUSD4 exhibited superior cytotoxicity over AGD owing to the induction of apoptotic cell death, mitochondrial membrane loss (ΔΨm), remarkable S-G2/M phase cell-cycle arrest and enhanced ROS generation in HT-29 cells. In conclusion, EUSD4 warrants further in-vivo antitumor testing under a set of stringent parameters against colorectal cancer.
Collapse
Affiliation(s)
- Pawan Devangan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Anamika Sharma
- Department of Biological Science, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Nitin Wadate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Atul Mourya
- Department of Biological Science, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India.
| |
Collapse
|
35
|
Yu J, Song L, Xu G, Li W, Liu S, Xie H, Tang J, Zhu J, Han XX. Monitoring of Propiolamide-Mediated Molecular Crosstalk between Ferroptosis and Apoptosis by Raman Spectroscopy. Anal Chem 2025; 97:5259-5265. [PMID: 40026133 DOI: 10.1021/acs.analchem.4c06968] [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: 03/04/2025]
Abstract
Inducing programmed cell death is an efficient strategy for cancer treatments, and a deep understanding of the molecular mechanisms underlying cell death pathways is of significance for the rational design of anticancer drugs. Herein, propiolamide-mediated crosstalk between ferroptosis and apoptosis is investigated. In situ monitoring of reactive oxygen species (ROS) formation and the structural changes of propiolamide compounds is achieved by Raman spectroscopy. The molecular mechanisms of propiolamides in inducing monooxygenase-mediated ROS generation and inhibiting GPX4 activities are revealed. Furthermore, the pro-ferroptotic and pro-apoptotic roles of the propiolamides containing terminal alkynes are verified. This study provides an in situ and label-free strategy for monitoring enzyme-drug interactions and their dynamics. It is a first attempt to study the structural basis of molecular crosstalk through two important enzymes in cell death. This study paves the way for designing novel drugs that are capable of triggering a synergistic contribution of multiple cell death forms to anticancer efficacy.
Collapse
Affiliation(s)
- Jiaheng Yu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Li Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Guangyang Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Wei Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Shiyi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Han Xie
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Jinping Tang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Jinyu Zhu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Xiao Xia Han
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
36
|
Xiang S, Yang DK. Aerial Yam Bulbils Protect Against APAP-Induced Hepatotoxicity by Inhibiting Oxidative Stress and Mitochondrial Dysfunction Through Nrf2 Activation. Nutrients 2025; 17:966. [PMID: 40289992 PMCID: PMC11944312 DOI: 10.3390/nu17060966] [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: 02/10/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND/OBJECTIVES The extract from aerial yam bulbils (AYB) contains various bioactive compounds, yet the mechanisms underlying its effects on APAP-induced liver injury need to be investigated further. This study sought to pursue the effects of AYB extract and the potential mechanisms involved in mitigating APAP-induced hepatotoxicity. METHODS TIB-73 cells were pretreated with AYB extract (10, 20, and 40 μg/mL) for 24 h and treated with APAP for 24 h to induce cytotoxicity. RESULTS Analysis of apoptosis-related proteins revealed that AYB extract exerts anti-apoptotic effects and inhibiting the MAPK signaling pathways, thereby reducing apoptotic cell death. Additionally, AYB extract significantly suppressed ROS overproduction by enhancing the expression of endogenous antioxidants and reducing the endoplasmic reticulum (ER) stress in APAP-treated cells, indicating that AYB extract inhibits APAP-induced oxidative stress. AYB extract effectively preserved mitochondrial membrane potential (MMP), maintained mitochondrial function-related genes, reduced mitochondrial oxidative stress, and mitigated mitochondrial damage, thereby preserving mitochondrial integrity. Additionally, AYB extract activated the Nrf2-related signaling pathway through nuclear translocation, leading to the upregulation of downstream antioxidative target genes. Diosgenin, a compound with known antioxidant properties and hepatoprotective effects, was identified in significant quantities in the AYB extract, suggesting that it may contribute to the observed hepatoprotective effects. CONCLUSIONS Overall, these findings demonstrate that AYB extract, with its antioxidative properties, effectively protects TIB-73 cells from APAP-induced liver injury.
Collapse
Affiliation(s)
- Siyu Xiang
- Department of Veterinary Pharmacology & Toxicology, College of Veterinary Medicine, Jeonbuk National University, Specialized Campus, 79 Gobong-ro, Iksan 54596, Jeollabuk-do, Republic of Korea;
| | - Dong Kwon Yang
- Department of Veterinary Pharmacology & Toxicology, College of Veterinary Medicine, Jeonbuk National University, Specialized Campus, 79 Gobong-ro, Iksan 54596, Jeollabuk-do, Republic of Korea;
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Republic of Korea
| |
Collapse
|
37
|
Zhao X, Zhang H, Liu Y, Li L, Wei H. Study on the metastatic mechanism of LINC00115 in adenocarcinoma of the Esophagogastric junction. Hum Mol Genet 2025; 34:492-511. [PMID: 39807637 DOI: 10.1093/hmg/ddae193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 10/16/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Adenocarcinoma of the esophagogastric junction (AEG) is a common and deadly cancer, and an in-depth investigation of its molecular mechanisms of metastasis is crucial for discovering new therapeutic targets. This study explores the role of the long non-coding RNA (lncRNA) LINC00115 in AEG metastasis and its underlying mechanisms. Through the analysis of 108 pairs of AEG cancer tissues and matched adjacent tissues, we found a significant upregulation of LINC00115 in AEG tissues, closely associated with TNM staging and lymph node metastasis. Utilizing cell counting kit-8 (CCK-8) assays, colony formation experiments, wound healing assays, flow cytometry for apoptosis and cell cycle analysis, and Transwell assays, we have confirmed that LINC00115 significantly promotes proliferation, migration, and invasion of AEG cells in vitro. Animal experiments further validate the role of LINC00115 in promoting tumor growth and metastasis in vivo. Additionally, our nuclear-cytoplasmic fractionation experiments and RNA fluorescence in situ hybridization (FISH) reveal that LINC00115, along with its interacting protein KH-Type splicing regulatory protein (KHSRP), predominantly localizes to the cell nucleus. By conducting RNA pull-down assays and mass spectrometry (MS) analysis, we have identified a direct interaction between LINC00115 and KHSRP protein and further determined their binding sites through catRAPID and ENCORI databases. This study provides evidence of LINC00115 as a novel biomarker and potential therapeutic target for AEG and offers a fresh perspective on understanding the molecular mechanisms of AEG metastasis.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Gastroenterology, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
| | - Haifeng Zhang
- Department of Thoracic Surgery, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
| | | | - Li Li
- Department of Thoracic Surgery, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
- Department of Thoracic Surgery, Huaihe Hospital of Henan University/Henan University School of Nursing and Health, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
| | - Haitao Wei
- Department of Thoracic Surgery, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng City, Henan Province, China
| |
Collapse
|
38
|
Yang D, Peng D, Zhou Y, Qiang Z, Wan L, Fan X, Meng Y, Xu G, Meng Y. Alpha-Momorcharin, a type I ribosome inactivating protein, induced apoptosis of hepatocellular carcinoma SK-HEP-1 cells through mitochondrial pathway. Nat Prod Res 2025; 39:1128-1138. [PMID: 38126176 DOI: 10.1080/14786419.2023.2295915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023]
Abstract
Alpha-Momorcharin (α-MMC), as one of the most important type I RIPs, has been reported to exert inhibitory effects against various tumour cells through its N-glycosidase activity. The present study was designed to propose an efficient purification strategy and explored its mechanism of apoptosis signalling pathway against human liver cancer cells SK-Hep-1. α-MMC can be successfully obtained by our purification strategy combining ion-exchange and gel-filtration chromatography. The functional studies revealed that α-MMC obviously increased the level of ROS and apoptosis rate, induced cell cycle arrest in the G1 phase, and depolarised MMP of SK-Hep-1 cells. To further confirm whether α-MMC could induce mitochondria involved apoptosis, we found that PARP-1, Caspase-3, Caspase-9, and BCL-2 were downregulated upon α-MMC. Taken together, these results suggested that this natural purified α-MMC can induce apoptosis involved mitochondria and may serve as a potential novel therapeutic drug in the treatment of human liver cancer in the future.
Collapse
Affiliation(s)
- Di Yang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Di Peng
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Yiping Zhou
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Sichuan Provincial People's Hospital Jinniu Hospital, Chengdu, Sichuan, China
| | - Zihao Qiang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Li Wan
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiang Fan
- Key Laboratory of Bio-resources and Eco-environment Ministry of Education/Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Yanfa Meng
- Key Laboratory of Bio-resources and Eco-environment Ministry of Education/Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Ge Xu
- The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, Sichuan, China
| | - Yao Meng
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
39
|
Wang Y, Su X, Wang Q, Zhang L, Yu Y, Zhao Y, Liu Z. Bisphenol A exposure enhances proliferation and tumorigenesis of papillary thyroid carcinoma through ROS generation and activation of NOX4 signaling pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117946. [PMID: 40014988 DOI: 10.1016/j.ecoenv.2025.117946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
As a prevalent industrial material and component of consumer products, bisphenol A (BPA) is linked to hormone homeostasis disruption and potential carcinogenicity. However, the precise mechanisms through which BPA contributes to thyroid carcinogenesis, especially in papillary thyroid carcinoma (PTC), are not fully understood. This study investigates how BPA boosts the proliferation and tumorigenic characteristics of thyroid cells. BPA exposure significantly increased cell proliferation in a duration-dependent manner at a concentration of 0.5 μM, which is slightly higher than human exposure levels. Therefore, this study utilized BPA treatment concentrations of 0.1 µM and 0.5 µM. BPA augmented the invasiveness of PTC cells with a dependency on both dosage and temporal factors. RNA-seq and gene expression analysis from normal human thyroid follicular epithelial cells suggested that BPA upregulated genes related to oxidative stress and thyroid cancer. Concurrently, our study revealed significant upregulation of NOX4 in thyroid tumors compared to normal thyroid tissues, with higher expression levels observed in advanced carcinomas by analyses of the TCGA database. BPA induces the upregulation of NOX4 in human thyroid cells, thereby triggering the activation of MAPK and PI3K/AKT pathways. In xenograft models, BPA treatment resulted in increased tumor size and Ki-67 proliferation index, accompanied by upregulated NOX4 expression. Additionally, BPA exposure led to higher levels of free triiodothyronine (FT3), indicating thyroid hormone disruption. Mechanistically, BPA activates the MAPK and PI3K/AKT pathways via NOX4, leading to increased ROS production and cell proliferation. This was further demonstrated through the use of ROS scavenger treatment and si-NOX4, which showed that BPA stimulates ROS generation by activating NOX4/MAPK and NOX4/PI3K/AKT pathways in thyroid cells. This finding enhances our understanding of the pathogenesis of PTC related to BPA exposure and highlights the necessity for rigorous health risk assessments regarding BPA exposure.
Collapse
Affiliation(s)
- Yi Wang
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xuling Su
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qianqian Wang
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Likun Zhang
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yaling Yu
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yiwei Zhao
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhiyan Liu
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
40
|
Wurlina W, Meles DK, Mustofa I, Khairullah AR, Putra DMS, Suwasanti N, Akintunde AO, Utama S, Mulyati S, Wasito W, Raissa R, Ahmad RZ, Julaeha J, Ekawasti F. Alkaloid fraction of Achyranthes aspera Linn triggers breast cancer apoptosis in mice ( Mus musculus) model. Open Vet J 2025; 15:1279-1288. [PMID: 40276193 PMCID: PMC12017709 DOI: 10.5455/ovj.2025.v15.i3.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 02/15/2025] [Indexed: 04/26/2025] Open
Abstract
Background Breast cancer affects women of various ages, and its recurrence is a significant cause of death. The search for potent anticancer compounds of herbal origin with well-defined mechanisms of action is an essential focus of current research. Aim This study aimed to investigate the effects of alkaloids in Achyranthes aspera Linn (AAL) leaf extract on necrosis, apoptosis, and related molecular markers, namely, cyclin-dependent kinase 1, Bcl-2 associated X-protein (Bax), rat sarcoma virus (Ras), cytochrome (Cyt) c, and apoptotic activating factor-1 (Apaf-1), in mice models. Methods Thirty mice with breast cancer were randomly divided into five groups. The negative control group only received distilled water daily. Mice in the positive control group (PCG) were administered methotrexate (15 mg/Kg) daily. The T1, T2, and T3 groups received oral orally at 75, 100, and 125 mg/Kg body weight daily for 30 days, respectively. On day 31, all mice were euthanized for the preparation of histological specimens of the mammary glands. The negative control group had the lowest number of apoptotic cells, Apaf-1, Cyt C, and Bax expression, and the highest number of viable cancer cells and Ras expression. Results The percentages of necrotic cells and breast cancer-expressed CDK-1 were not significantly (p > 0.05) different among groups. The percentage of apoptotic cells, Apaf-1, and Cyt c, was highest in T3. Conversely, the percentage of viable cells and breast cancer-expressing Ras was lowest in T3. Conclusion Treatment with 125 mg/Kg AAL suppressed cancer cell growth in breast cancer-bearing mice. Further research is necessary to determine the complete signaling mechanism.
Collapse
Affiliation(s)
- Wurlina Wurlina
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Dewa Ketut Meles
- Division of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Imam Mustofa
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Aswin Rafif Khairullah
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | | | - Niluh Suwasanti
- Department of Clinical Pathology, Faculty of Medicine, Universitas Katolik Widya Mandala Surabaya, Surabaya, Indonesia
| | - Adeyinka Oye Akintunde
- Department of Agriculture and Industrial Technology, Babcock University, Ilishan Remo State, Nigeria
| | - Suzanita Utama
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Sri Mulyati
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Wasito Wasito
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Ricadonna Raissa
- Department of Pharmacology, Faculty of Veterinary Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Riza Zainuddin Ahmad
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Julaeha Julaeha
- Research Center for Preclinical and Clinical Medicine, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Fitrine Ekawasti
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| |
Collapse
|
41
|
Kausar R, Nguyen NTT, Le TPH, Kim JH, Lee SY. Inhibition of HDAC6 elicits anticancer effects on head and neck cancer cells through Sp1/SOD3/MKP1 signaling axis to downregulate ERK phosphorylation. Cell Signal 2025; 127:111587. [PMID: 39755348 DOI: 10.1016/j.cellsig.2024.111587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
Oxidative stress caused by reactive oxygen species (ROS) and superoxides is linked to various cancer-related biological events. Extracellular superoxide dismutase (SOD3), an antioxidant enzyme that removes superoxides, contributes to redox homeostasis and has the potential to regulate tumorigenesis. Histone deacetylase 6 (HDAC6), a major HDAC isoform responsible for mediating the deacetylation of non-histone protein substrates, also plays a role in cancer progression. In this study, we examined the potential effects of HDAC6 inhibition on SOD3 expression in head and neck cancer (HNC) cells and its impact on cell proliferation, which remains unaddressed. We found that functional inactivation of HDAC6, through the use of chemical inhibitors such as tubastatin A (TubA), gene knockdown, or overexpression of an inactive mutant, strongly upregulated protein and mRNA levels of SOD3 in HNC cell lines FaDu and Detroit562. Mechanistically, TubA induced acetylation of the transcription factor Sp1 at Lys703, which consequently enhanced its binding to the SOD3 proximal promoter region and increased SOD3 expression. An acetylation-defective Sp1 mutant (K703R) was much less effective in inducing SOD3 expression compared to wild-type Sp1. TubA reduced intracellular ROS and superoxide levels, and this antioxidative effect was attenuated in SOD3 knockdown cells. Similar to the changes in ROS levels, HDAC6 inhibition as well as SOD3 overexpression suppressed cell proliferation and the stimulatory phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), whereas SOD3 knockdown produced opposite effects in both resting and TubA-treated conditions. In addition, SOD3 overexpression prevented ROS-induced ERK1/2 phosphorylation and enhanced the protein stability of mitogen-activated protein kinase phosphatase 1 (MKP1), thereby counteracting ERK1/2 phosphorylation. We further showed that SOD3-mediated ERK1/2 dephosphorylation was moderated in MKP1 knockdown cells. Collectively, these results suggest that HDAC6 inhibition elicits anticancer effects on HNC cells by promoting Sp1 acetylation-dependent SOD3 upregulation, leading to MKP1 stabilization and subsequent ERK1/2 inactivation.
Collapse
Affiliation(s)
- Rukhsana Kausar
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Gyeonggi 16499, Republic of Korea
| | - Nga Thi Thanh Nguyen
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi 16499, Republic of Korea
| | - Truc Phan Hoang Le
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi 16499, Republic of Korea
| | - Jae Hyung Kim
- Department of Anesthesiology and Pain Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi 18450, Republic of Korea
| | - Sang Yoon Lee
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Gyeonggi 16499, Republic of Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi 16499, Republic of Korea.
| |
Collapse
|
42
|
Wu M, Wu B, Huang X, Wang Z, Zhu M, Zhu Y, Yu L, Liu J. Inhibition of the FEN1-PBX1 axis elicits cellular senescence in breast cancer via the increased intracellular reactive oxygen species levels. J Transl Med 2025; 23:248. [PMID: 40022092 PMCID: PMC11871692 DOI: 10.1186/s12967-025-06216-9] [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: 09/18/2024] [Accepted: 02/10/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Cellular senescence is a state of irreversible cell growth arrest. As such, senescence induction is viewed as an efficacious countermeasure in cancer treatment. Flap endonuclease 1 (FEN1) has been reported to participate in tumor growth, metastasis and immunomodulation. However, the role of FEN1 in cellular senescence of breast cancer and its molecular mechanism remains unclear. METHODS In vitro assessments of breast cancer cell senescence and apoptosis were conducted using CCK-8 assay, cell cycle assay, senescence-associated β-galactosidase (SA-β-gal) staining, and cleaved caspase-3 staining. Western blot, dihydroethidium (DHE) staining, RNA-sequencing, quantitative real-time polymerase chain reaction (qRT-PCR), rescue experiments, and dual-luciferase reporter assay were performed to explore the potential target of FEN1. Co-Immunoprecipitation (Co-IP), Chromatin immunoprecipitation (ChIP)-qPCR assay, and immunostaining were used to evaluate the interaction between FEN1 and Pre-B-cell leukemia homeobox transcription factor 1 (PBX1). A xenograft mouse model was employed to validate the effect of FEN1 on breast cancer cell senescence and apoptosis. RESULTS Functional analysis demonstrated that FEN1 suppressed both senescence and apoptosis of breast cancer cells in vitro, while in vivo experiments demonstrated moderate therapeutic effects. Further studies indicated that FEN1 deficiency promoted the aforementioned effects by increasing intracellular reactive oxygen species (ROS) levels. RNA-sequencing and qRT-PCR assays revealed that FEN1 knockdown enhanced the expressions of several senescence-associated secretory phenotype (SASP) factors and resulted in decreased PBX1 level. The rescue experiments by PBX1 overexpression verified that PBX1 mediated the senescence and apoptosis of breast cancer cells induced by FEN1 inhibition. In detail, FEN1 downregulation inhibited the transcription activity of PBX1, which was partially restored by itself overexpression. Of note, FEN1 directly interacted with PBX1. Furthermore, immunostaining illustrated the colocalization of FEN1 and PBX1 in breast cancer cells and tissues. In our local breast cancer cohort, a positive correlation was identified between the expression levels of FEN1 and PBX1. CONCLUSIONS Knockdown of FEN1 facilitates breast cancer cell senescence through PBX1 down-regulation mediating increase in intracellular ROS levels. This study reveals FEN1 as a negative regulator of cellular senescence and provides support for pro-senescence cancer therapy. Given that FEN1 knockdown exhibited only moderate in vivo effects, these findings underscore the necessity of combining it with senolytic therapy to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Min Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| | - Benmeng Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoshan Huang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Zirui Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Miaolin Zhu
- Department of Pathology, Jiangsu Cancer Hospital, Nanjing, 210018, China
| | - Yaqin Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Lin Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Jingjing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
43
|
Gollowitzer A, Pein H, Rao Z, Waltl L, Bereuter L, Loeser K, Meyer T, Jafari V, Witt F, Winkler R, Su F, Große S, Thürmer M, Grander J, Hotze M, Harder S, Espada L, Magnutzki A, Gstir R, Weinigel C, Rummler S, Bonn G, Pachmayr J, Ermolaeva M, Harayama T, Schlüter H, Kosan C, Heller R, Thedieck K, Schmitt M, Shimizu T, Popp J, Shindou H, Kwiatkowski M, Koeberle A. Attenuated growth factor signaling during cell death initiation sensitizes membranes towards peroxidation. Nat Commun 2025; 16:1774. [PMID: 40000627 PMCID: PMC11861335 DOI: 10.1038/s41467-025-56711-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Cell death programs such as apoptosis and ferroptosis are associated with aberrant redox homeostasis linked to lipid metabolism and membrane function. Evidence for cross-talk between these programs is emerging. Here, we show that cytotoxic stress channels polyunsaturated fatty acids via lysophospholipid acyltransferase 12 into phospholipids that become susceptible to peroxidation under additional redox stress. This reprogramming is associated with altered acyl-CoA synthetase isoenzyme expression and caused by a decrease in growth factor receptor tyrosine kinase (RTK)-phosphatidylinositol-3-kinase signaling, resulting in suppressed fatty acid biosynthesis, for specific stressors via impaired Akt-SREBP1 activation. The reduced availability of de novo synthesized fatty acids favors the channeling of polyunsaturated fatty acids into phospholipids. Growth factor withdrawal by serum starvation mimics this phenotype, whereas RTK ligands counteract it. We conclude that attenuated RTK signaling during cell death initiation increases cells' susceptibility to oxidative membrane damage at the interface of apoptosis and alternative cell death programs.
Collapse
Affiliation(s)
- André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Helmut Pein
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Zhigang Rao
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Lorenz Waltl
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Leonhard Bereuter
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Konstantin Loeser
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Tobias Meyer
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Vajiheh Jafari
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Finja Witt
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - René Winkler
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
- Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916, Badalona, Spain
| | - Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Silke Große
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Maria Thürmer
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Julia Grander
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Madlen Hotze
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Sönke Harder
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Lilia Espada
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Alexander Magnutzki
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Ronald Gstir
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Christina Weinigel
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Silke Rummler
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Günther Bonn
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Johanna Pachmayr
- Institute of Pharmacy, Paracelsus Medical University, 5020, Salzburg, Austria
| | - Maria Ermolaeva
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Takeshi Harayama
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur - CNRS UMR7275 - Inserm U1323, 06560, Valbonne, France
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
| | - Regine Heller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Kathrin Thedieck
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
- Department Metabolism, Senescence and Autophagy, Research Center One Health Ruhr, University Alliance Ruhr & University Hospital Essen, University Duisburg-Essen, 45141, Essen, Germany
- Freiburg Materials Research Center FMF, Albert-Ludwigs-University of Freiburg, 79104, Freiburg, Germany
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
- German Cancer Consortium (DKTK), partner site Essen/Duesseldorf, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147, Essen, Germany
| | - Michael Schmitt
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Institute of Microbial Chemistry, Tokyo 141-0021, Japan
| | - Jürgen Popp
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Hideo Shindou
- Department of Lipid Life Science, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Marcel Kwiatkowski
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria.
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany.
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria.
| |
Collapse
|
44
|
Tao Y, Zhuang W, Fan W, Zhou L, Fan L, Qin H, Zhu Y. Dual-functional silver nanoparticle-enhanced ZnO nanorods for improved reactive oxygen species generation and cancer treatment. iScience 2025; 28:111858. [PMID: 40017508 PMCID: PMC11867527 DOI: 10.1016/j.isci.2025.111858] [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: 05/05/2024] [Revised: 08/22/2024] [Accepted: 01/17/2025] [Indexed: 03/01/2025] Open
Abstract
Recent advancements in sonodynamic therapy (SDT) for cancer treatment have highlighted the potential of enhancing reactive oxygen species (ROS) generation and improving therapeutic outcomes. This study introduces zinc oxide (ZnO) nanorods (NRs) in situ loaded with silver nanoparticles (ZnO@Ag NRs), designed to optimize ROS production under ultrasound irradiation and offer significant advantages in tumor specificity and biosafety. The transmission electron microscopy and elemental mapping confirmed the consistent size and monodispersed Ag nanoparticle for ZnO@Ag NR. Sonodynamic properties showed that ZnO@Ag NRs produce higher singlet oxygen and hydroxyl radicals under ultrasound. In vitro studies demonstrated excellent biocompatibility and enhanced cell-killing effects of ZnO@Ag NRs on CT-26 cells, while in vivo results confirmed its superior anti-tumor efficacy and biosafety. Furthermore, the ZnO@Ag NRs' antibacterial properties were also confirmed, suggesting additional benefits in treating cancers associated with bacterial infections. Collectively, these findings establish ZnO@Ag NRs as a potent and safe agent for ultrasound-driven cancer therapy.
Collapse
Affiliation(s)
- Yichao Tao
- School of Medicine, Nantong University, Nantong 226001, China
- Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
- Zhangjiagang Hospital of Traditional Chinese Medicine, Suzhou 215600, China
| | - Wenbin Zhuang
- School of Medicine, Nantong University, Nantong 226001, China
- Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Wensi Fan
- Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Longxiang Zhou
- Department of General Surgery, Jinshan Central Hospital Affiliated to Shanghai University of Medicine & Health Sciences, No.147, Jiankang Road, Shanghai 201599, China
| | - Lihong Fan
- School of Medicine, Nantong University, Nantong 226001, China
- Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Huanlong Qin
- School of Medicine, Nantong University, Nantong 226001, China
- Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yefei Zhu
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
45
|
Jin S, Qian W, Suo R, Li Y, Ling H, Li S, Deng K, Wei Y, Wu B, Chen H. Radical-triggered ring-opening of aminocyclopropane for detection of hydroxyl radicals in living cells. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:1892-1899. [PMID: 39925080 DOI: 10.1039/d4ay02150a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Hydroxyl radicals (˙OH), highly reactive oxygen species involved in oxidative stress and cancer therapy, are challenging to detect intracellularly due to their short lifetime, low concentration, and high reactivity. To address this, a novel ˙OH-specific fluorescent probe, CC-7, was developed by integrating an aminocyclopropane group into a coumarin derivative. This design was inspired by the radical-mediated ring-opening of aminocyclopropanes in synthetic chemistry. The ring-opening reaction triggered by ˙OH in CC-7 produces a significant "Fluorescence-ON" response with a 10-fold increase in intensity, demonstrating high selectivity for ˙OH over other reactive oxygen species. CC-7 effectively visualized intracellular ˙OH, distinguished between normal (HEK-293T) and cancer cells (4T1), and monitored ˙OH generated by chemotherapeutic agents like doxorubicin and cisplatin. This study highlights CC-7 as a powerful tool for selectively detecting ˙OH in living cells, with potential applications in investigating oxidative stress-related diseases and monitoring cancer therapy.
Collapse
Affiliation(s)
- Shiqi Jin
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China.
- School of Chemistry and Chemical Engineering, Hubei University, Wuhan 430071, P. R. China.
| | - Wang Qian
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China.
| | - Ruiyang Suo
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China.
| | - Yi Li
- School of Chemistry and Chemical Engineering, Hubei University, Wuhan 430071, P. R. China.
| | - Huan Ling
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China.
| | - Shuqi Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China.
| | - Kai Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China.
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China.
| | - Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China.
| | - Huaixia Chen
- School of Chemistry and Chemical Engineering, Hubei University, Wuhan 430071, P. R. China.
| |
Collapse
|
46
|
Shani MY, Ashraf MY, Ramzan M, Khan Z, Batool N, Gul N, Bauerle WL. Unveiling Drought Tolerant Cotton Genotypes: Insights from Morpho-Physiological and Biochemical Markers at Flowering. PLANTS (BASEL, SWITZERLAND) 2025; 14:616. [PMID: 40006874 PMCID: PMC11859814 DOI: 10.3390/plants14040616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Drought stress substantially restricts cotton growth, decreasing cotton production potential worldwide. This study evaluated cotton genotypes at the flowering stage to identify drought-resilient genotypes under moderate and severe drought conditions using physio-morphic and biochemical markers. Five genotypes were examined in a completely randomized design with three replicates across three treatments. Growth and biochemical traits were measured after 14 days of drought stress. The Multi-trait Genotype-Ideotype Distance Index (MGIDI) identified the most drought-tolerant genotypes. Severe drought had a pronounced negative effect on growth and biochemical traits, followed by moderate drought. Among the genotypes, FH-912 exhibited the strongest resilience, with significant increases in proline, peroxidase, catalase, and total chlorophyll. In contrast, chlorophyll a and transpiration rates were largely unaffected. Genotypes VH-351, VH-281, and GH-99 showed moderate drought tolerance, while FH-556 was highly sensitive to water stress. Statistical analyses, including ANOVA, PCA, and heatmaps, confirmed FH-912's superior performance under drought stress. The drought-resilient genotype, FH-912, holds promise for breeding drought-tolerant cotton varieties to sustain cotton productivity in water-limited environments, especially in drought-prone regions.
Collapse
Affiliation(s)
- Muhammad Yousaf Shani
- Plant Breeding and Genetics Division, Nuclear Institute for Agriculture and Biology, Jhang Road, P.O. Box 128, Faisalabad 38000, Pakistan;
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nuclear Institute for Agriculture and Biology/College (NIAB-C), Islamabad 45650, Pakistan;
| | - Muhammad Yasin Ashraf
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nuclear Institute for Agriculture and Biology/College (NIAB-C), Islamabad 45650, Pakistan;
| | - Muhammad Ramzan
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nuclear Institute for Agriculture and Biology/College (NIAB-C), Islamabad 45650, Pakistan;
| | - Zafran Khan
- Department of Plant Breeding and Genetics, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan; (Z.K.); (N.G.)
| | - Nimra Batool
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore 54000, Pakistan;
| | - Nimra Gul
- Department of Plant Breeding and Genetics, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan; (Z.K.); (N.G.)
| | - William L. Bauerle
- Department of Horticulture and Landscape Architecture, Colorado State University, Fort Collins, CO 80523, USA;
| |
Collapse
|
47
|
Li X, Wang X, Chen G, Tian B. Application trends of hydrogen-generating nanomaterials for the treatment of ROS-related diseases. Biomater Sci 2025; 13:896-912. [PMID: 39807026 DOI: 10.1039/d4bm01450b] [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: 01/16/2025]
Abstract
Reactive oxygen species (ROS) play essential roles in both physiological and pathological processes. Under physiological conditions, appropriate amounts of ROS play an important role in signaling and regulation in cells. However, too much ROS can lead to many health problems, including inflammation, cancer, delayed wound healing, neurodegenerative diseases (such as Parkinson's disease and Alzheimer's disease), and autoimmune diseases, and oxidative stress from excess ROS is also one of the most critical factors in the pathogenesis of cardiovascular and metabolic diseases such as atherosclerosis. Hydrogen gas effectively removes ROS from the body due to its good antioxidant properties, and hydrogen therapy has become a promising gas therapy strategy due to its inherent safety and stability. The combination of nanomaterials can achieve targeted delivery and effective accumulation of hydrogen, and has some ameliorating effects on diseases. Herein, we summarize the use of hydrogen-producing nanomaterials for the treatment of ROS-related diseases and talk about the prospects for the treatment of other ROS-induced disease models, such as acute kidney injury.
Collapse
Affiliation(s)
- Xiaobing Li
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Xuezhu Wang
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Guifang Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Bo Tian
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
48
|
Li J, Liu W, Zhang J, Sun C. The Role of Mitochondrial Quality Control in Liver Diseases: Dawn of a Therapeutic Era. Int J Biol Sci 2025; 21:1767-1783. [PMID: 39990657 PMCID: PMC11844277 DOI: 10.7150/ijbs.107777] [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: 11/28/2024] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
The liver is a vital metabolic organ that detoxifies substances, produces bile, stores nutrients, and regulates versatile metabolic processes. Maintaining normal liver cell function requires the prompt and delicate modulation of mitochondrial quality control (MQC), which encompasses a spectrum of processes such as mitochondrial fission, fusion, biogenesis, and mitophagy. Recent studies have shown that disruptions to this homeostatic status are closely linked to the advent and progression of a variety of acute and chronic liver diseases, including but not limited to alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease. However, the explicit mechanisms by which mitochondrial dysfunction impacts inflammatory pathways and cell death in the context of liver diseases remain unclear. In this narrative review, we provide a detailed description of MQC, analyze the mechanisms underpinning mitochondrial dysfunction induced by different detrimental insults, and further elucidate how imbalanced/disrupted MQC promotes the progression and aggravation of liver diseases, ultimately shedding light on the mitochondrion-centric therapeutic strategies for these pathophysiological entities.
Collapse
Affiliation(s)
- Jia Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Wenqin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Jie Zhang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Chao Sun
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
- Department of Gastroenterology, Tianjin Medical University General Hospital Airport Hospital, East Street 6, Tianjin Airport Economic Area, Tianjin 300308, China
| |
Collapse
|
49
|
Zheng S, Piao Y, Jung SN, Oh C, Lim MA, Nguyen Q, Shen S, Park SH, Cui S, Piao S, Kim YI, Kim JW, Won HR, Chang JW, Shan Y, Liu L, Koo BS. Gene Expression Alteration by Non-thermal Plasma-Activated Media Treatment in Radioresistant Head and Neck Squamous Cell Carcinoma. Clin Exp Otorhinolaryngol 2025; 18:73-87. [PMID: 39757757 PMCID: PMC11917201 DOI: 10.21053/ceo.2024.00238] [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: 08/14/2024] [Accepted: 01/05/2025] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) exhibits high recurrence rates, particularly in cases of radioresistant HNSCC (RR-HNSCC). Non-thermal plasma (NTP) therapy effectively suppresses the progression of HNSCC. However, the therapeutic mechanisms of NTP therapy in treating RR-HNSCC are not well understood. In this study, we explored the regulatory role of NTP in the RR-HNSCC signaling pathway and identified its signature genes. METHODS After constructing two RR-HNSCC cell lines, we prepared cell lysates from cells treated or not treated with NTP-activated media (NTPAM) and performed RNA sequencing to determine their mRNA expression profiles. Based on the RNA sequencing results, we identified differentially expressed genes (DEGs), followed by a bioinformatics analysis to identify candidate molecules potentially associated with NTPAM therapy for RR-HNSCC. RESULTS NTPAM reduced RR-HNSCC cell viability in vitro. RNA sequencing results indicated that NTPAM treatment activated the reactive oxygen species (ROS) pathway and induced ferroptosis in RR-HNSCC cell lines. Among the 1,924 genes correlated with radiation treatment, eight showed statistical significance in both the cell lines and The Cancer Genome Atlas (TCGA) cohort. Only five genes-ABCC3, DUSP16, PDGFB, RAF1, and THBS1-showed consistent results between the NTPAM data sequencing and TCGA data. LASSO regression analysis revealed that five genes were associated with cancer prognosis, with a hazard ratio of 2.26. In RR-HNSCC cells, NTPAM affected DUSP16, PDGFB, and THBS1 as activated markers within 6 hours, and this effect persisted for 12 hours. Furthermore, enrichment analysis indicated that these three DEGs were associated with the extracellular matrix, transforming growth factor-beta, phosphoinositide 3-kinase/protein kinase B, and mesenchymal-epithelial transition factor pathways. CONCLUSION NTPAM therapy exerts cytotoxic effects in RR-HNSCC cell lines by inducing specific ROS-mediated ferroptosis. DUSP16, PDGFB, and THBS1 were identified as crucial targets for reversing the radiation resistance induced by NTPAM therapy, providing insights into the mechanisms and clinical applications of NTPAM treatment in RR-HNSCC.
Collapse
Affiliation(s)
- Sicong Zheng
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yudan Piao
- Dental Department, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seung-Nam Jung
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Chan Oh
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Mi Ae Lim
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - QuocKhanh Nguyen
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Shan Shen
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Se-Hee Park
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Shengzhe Cui
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Shuyu Piao
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Young Il Kim
- Department of Radiation Oncology, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Ji Won Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Ho-Ryun Won
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Jae Won Chang
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yujuan Shan
- Department of Nutrition, Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Lihua Liu
- Department of Nutrition, Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Bon Seok Koo
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
50
|
Varlamova EG, Gudkov SV, Turovsky EA. Differential effect of cerium nanoparticles on the viability, redox-status and Ca 2+-signaling system of cancer cells of various origins. Arch Biochem Biophys 2025; 764:110261. [PMID: 39645139 DOI: 10.1016/j.abb.2024.110261] [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: 08/18/2024] [Revised: 11/12/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The present study aims to understand the molecular mechanism underlying the therapeutic effect of cerium nanoparticles (CeNPs) in oncology. Cancer cells were treated with different concentrations of pure nanocerium of different sizes synthesized by laser ablation. Due to the not insignificant influence of surface defects and oxygen species on the ROS-modulating properties of cerium nanoparticles, the nanoparticles were not coated with surfactants or organic molecules during synthesis, which could potentially inhibit a number of pro-oxidative effects. Reactive oxygen species (ROS) production, expression of genes encoding redox-status proteins, selenoproteins and proteins regulating cell death and endoplasmic reticulum stress (ER-stress) were investigated as indicators of the molecular mechanism of cancer cell death. Studies were conducted on the effects of cerium nanoparticles on the Ca2+ signaling system of cancer cells of different origins. Mouse fibroblasts (L-929 cell line) were used as non-cancerous ("normal") cells for which a whole series of experiments were performed, and a comparative analysis of the effects of nanoceria. It was found that 75 nm-sized cerium nanoparticles did not affect the redox-status and ROS production of cancer cells. In fibroblast cells, however, this nanoparticle diameter led to a deterioration of the cellular redox status and ROS production in a wide range of nanoparticle concentrations. Larger nanoparticles (100 nm-sized and 160 nm-sized), on the other hand, showed a different effect on cancer cells of different origins. In mouse fibroblast L-929 cells, however, 100 nm-sized or 160 nm-sized CeNPs acted in a high concentration range to disrupt mitochondrial membrane potential and activate early apoptosis. High concentrations of CeNPs were required to increase ROS production, reduce redox-status and induce apoptosis in human A-172 glioblastoma cells compared to the hepatocellular carcinoma cell line HepG2 and the breast cancer cell line MCF-7. In the A-172 glioblastoma cells, ER-stress was also not activated and their Ca2+ signaling system was activated by a significantly higher concentration of CeNPs, which could also contribute to the formation of tolerance of this cancer cell line to nanoceria. The Ca2+ signaling system of mouse fibroblasts was found to be highly sensitive to activation by nanoceria and the cells produced Ca2+ signals with higher amplitude compared to A-172 and MCF-7 cells.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia
| | - Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilove st., 119991, Moscow, Russia
| | - Egor A Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia.
| |
Collapse
|