1
|
Wang J, Guo Q, He L, Song R, Du J, Zhou H, Hao Y, Yang X, Wang F, Li K, Li M, Yang Z, Sun L, Liu Z. A Nanoradiosensitizer Potentiates Tumor Radiotherapy through JFK Inhibition and Hypoxia Alleviation. NANO LETTERS 2025; 25:5435-5443. [PMID: 40125668 DOI: 10.1021/acs.nanolett.5c00677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Radiotherapy (RT) is a primary treatment for breast cancer, but its effectiveness is often compromised by hypoxia and intrinsic resistance mechanisms. The F-box protein JFK is overexpressed in breast cancer and is associated with reduced radiosensitivity, but specific JFK inhibitors are currently unavailable. Herein, we developed spherical nanoparticles (SNP-JC) designed to co-deliver small interfering RNA targeting JFK and catalase to the tumor, aiming to silence JFK and alleviate hypoxia to overcome RT resistance. Positron emission tomography imaging demonstrated that SNP-JC efficiently accumulated in the tumors. SNP-JC significantly increased DNA damage in tumor cells after RT and promoted the immunogenic cell death. The combination of SNP-JC and RT activated CD8+ T cells and elicited a robust antitumor immunity, resulting in suppressed primary tumor growth and reduced lung metastasis. Our findings demonstrate that a nanoplatform capable of simultaneously silencing JFK and mitigating hypoxia can enhance tumor radiosensitivity, improve antitumor efficacy, and prevent metastasis.
Collapse
Affiliation(s)
- Jianze Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Qianrui Guo
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Lin He
- Department of Biochemistry, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Rui Song
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Jinhong Du
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Haoyi Zhou
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Yameng Hao
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Xiujie Yang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Feng Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Kui Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Mo Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Zhi Yang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Luyang Sun
- Department of Biochemistry, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Zhaofei Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital and Institute, Beijing 100142, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Nuclear Medicine, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
2
|
Yu S, Wu J, Jing Y, Lin P, Lang L, Xiong Y, Chen W, Liu W, Sun C, Lu Y. Research trends in glioma chemoradiotherapy resistance: a bibliometric analysis (2003-2023). Front Oncol 2025; 15:1539937. [PMID: 39990688 PMCID: PMC11842341 DOI: 10.3389/fonc.2025.1539937] [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: 12/05/2024] [Accepted: 01/16/2025] [Indexed: 02/25/2025] Open
Abstract
Background Glioma is the most aggressive primary malignant tumor of the central nervous system, characterized by high recurrence rates and resistance to chemoradiotherapy, making therapeutic resistance a major challenge in neuro-oncology. Recent research emphasizes the role of the tumor microenvironment (TME) and immune modulation in glioma progression and resistance. Despite these advances, a comprehensive bibliometric analysis of research trends in glioma chemoradiotherapy resistance over the past two decades is lacking. This study aims to systematically evaluate the research landscape, identify emerging hotspots, and provide guidance for future investigations. Methods Articles on glioma chemoradiotherapy resistance published between 2003 and 2023 were retrieved from the Web of Science Core Collection, resulting in 4,528 publications. Bibliometric tools, including VOSviewer, CiteSpace, and R packages such as bibliometrix and ggplot2, were used to analyze co-authorship networks, keyword evolution, and citation bursts to identify collaboration patterns, thematic developments, and influential contributions. Results Publication output increased significantly between 2013 and 2022, peaking at 650 articles in 2022. Over 1,000 institutions from 88 countries contributed to this research. The United States, Switzerland, and Germany showed the highest citation impact, while China led in publication volume but demonstrated relatively lower citation influence. The research focus has shifted from traditional topics such as the "MGMT gene" to emerging areas including the "tumor microenvironment," "immune infiltration," and "nanoparticles." The androgen receptor was identified as a promising but underexplored therapeutic target. Conclusions Research on glioma chemoradiotherapy resistance has seen substantial growth, with increasing emphasis on immune modulation, the tumor microenvironment, and novel therapeutic targets such as the androgen receptor. This study represents the first comprehensive bibliometric analysis of this field, providing a detailed overview of research trends and potential directions for future studies. The findings highlight the need for strengthened international collaboration and multidisciplinary approaches to address the challenges of therapeutic resistance in glioma.
Collapse
Affiliation(s)
- Shishi Yu
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinya Wu
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuan Jing
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Ping Lin
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Lang Lang
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Yifan Xiong
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Wangzhong Chen
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenhua Liu
- Clinical Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changpeng Sun
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuntao Lu
- Nanfang hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Rusak A, Wiatrak B, Krawczyńska K, Górnicki T, Zagórski K, Zadka Ł, Fortuna W. Starting points for the development of new targeted therapies for glioblastoma multiforme. Transl Oncol 2025; 51:102187. [PMID: 39531784 PMCID: PMC11585793 DOI: 10.1016/j.tranon.2024.102187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/30/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive and lethal brain tumors, characterized by rapid growth, invasiveness, and resistance to standard therapies, including surgery, chemotherapy, and radiotherapy. Despite advances in treatment, GBM remains highly resistant due to its complex molecular mechanisms, including angiogenesis, invasion, immune modulation, and lipid metabolism dysregulation. This review explores recent breakthroughs in targeted therapies, focusing on innovative drug carriers such as nanoparticles and liposomes, and their potential to overcome GBM's chemo- and radioresistant phenotypes. We also discuss the molecular pathways involved in GBM progression and the latest therapeutic strategies, including immunotherapy and precision medicine approaches, which hold promise for improving clinical outcomes. The review highlights the importance of understanding GBM's genetic and molecular heterogeneity to develop more effective, personalized treatment protocols aimed at increasing survival rates and enhancing the quality of life for GBM patients.
Collapse
Affiliation(s)
- Agnieszka Rusak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland.
| | - Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, J. Mikulicza-Radeckiego 2 Street, Wroclaw 50-345, Poland.
| | - Klaudia Krawczyńska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland.
| | - Tomasz Górnicki
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland
| | - Karol Zagórski
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland
| | - Łukasz Zadka
- Division of Ultrastructural Research, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland; Department of Clinical Pharmacology, Wroclaw Medical University, Borowska 211a, Wroclaw 50-556, Poland.
| | - Wojciech Fortuna
- Department of Neurosurgery, Wroclaw Medical University, Borowska 213St, Wroclaw 50-556, Poland.
| |
Collapse
|
4
|
Ge Y, Ge Z, Tian F, Tai X, Chen D, Sun S, Shi Z, Yin J, Wei G, Li D, Wang L, Xu W, Tong M, Liu F, Zhao L, Qian X, Ge X. Sulforaphane potentiates the efficacy of chemoradiotherapy in glioblastoma by selectively targeting thioredoxin reductase 1. Cancer Lett 2024; 611:217429. [PMID: 39725145 DOI: 10.1016/j.canlet.2024.217429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Chemoradiotherapy is a conventional treatment modality for patients with glioblastoma (GBM). However, the efficacy of this approach is significantly hindered by the development of therapeutic resistance. The thioredoxin system, which plays a crucial role in maintaining redox homeostasis, confers protection to cancer cells against apoptosis induced by chemoradiotherapy. Herein, we demonstrate that sulforaphane (SFN), an isothiocyanate phytochemical with anti-cancer effects, inhibits the activity of thioredoxin reductase 1 (TrxR1) through covalent conjugation with residues C59/64/497&U498. This inhibition of TrxR1 leads to the accumulation of reactive oxygen species (ROS), thereby enhancing chemoradiotherapy-induced apoptosis in GBM cells. Furthermore, SFN-induced ROS accumulation facilitates the polarization of M1-like macrophages, which synergistically sensitize GBM tumors to chemoradiotherapy. In conclusion, our study unveils that SFN has potential benefits in improving the effect of chemoradiotherapy and prognosis for GBM patients by targeting TrxR1.
Collapse
Affiliation(s)
- Yuqian Ge
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Zehe Ge
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Fuwei Tian
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiaoyu Tai
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Dongyin Chen
- Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Shuhong Sun
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Zhumei Shi
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jianxing Yin
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Guining Wei
- Department of Pharmacology, Guangxi Institute of Chinese Medicine & Pharmaceutical Science, Nanning, 530022, China
| | - Dongmei Li
- Department of Pharmacology, Guangxi Institute of Chinese Medicine & Pharmaceutical Science, Nanning, 530022, China
| | - Lude Wang
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Wenxia Xu
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Minfeng Tong
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Fang Liu
- Department of Neurosurgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China
| | - Lin Zhao
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Xu Qian
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital and Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 21009, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Xin Ge
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
5
|
Li Y, Liu C, Fang B, Chen X, Wang K, Xin H, Wang K, Yang SM. Ferroptosis, a therapeutic target for cardiovascular diseases, neurodegenerative diseases and cancer. J Transl Med 2024; 22:1137. [PMID: 39710702 DOI: 10.1186/s12967-024-05881-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The identification of ferroptosis represents a pivotal advancement in the field of cell death research, revealing an entirely novel mechanism of cellular demise and offering new insights into the initiation, progression, and therapeutic management of various diseases. Ferroptosis is predominantly induced by intracellular iron accumulation, lipid peroxidation, or impairments in the antioxidant defense system, culminating in membrane rupture and consequent cell death. Studies have associated ferroptosis with a wide range of diseases, and by enhancing our comprehension of its underlying mechanisms, we can formulate innovative therapeutic strategies, thereby providing renewed hope for patients.
Collapse
Affiliation(s)
- Yinghui Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Cuiyun Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bo Fang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xinzhe Chen
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Kai Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China.
| | - Kun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Su-Min Yang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
6
|
Zang W, Liu Y, Zheng J, Huang Y, Chen L, Li C, Zhao J, Zhou Q, Xu Y, Wang Z, Cao Y, Zhang W, Li J, Lu Y. LAT4 drives temozolomide induced radiotherapy resistance in glioblastoma by enhancing mTOR pathway activation. Cancer Cell Int 2024; 24:407. [PMID: 39696435 DOI: 10.1186/s12935-024-03590-0] [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: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) represents the most prevalent form of primary malignant tumor within the central nervous system. The emergence of resistance to radiotherapy and chemotherapy represents a significant impediment to advancements in glioma treatment. METHODS We established temozolomide (TMZ)-resistant GBM cell lines by chronically exposing U87MG cell lines to TMZ, and dimethyl sulfoxide (DMSO) was used as placebo control. In vivo and in vitro experiments verified the resistance of resistant cells to chemotherapy and radiotherapy. LAT4 was identified by transcriptomics to be associated with GBM treatment resistance and relapse. The relationship between LAT4 and mTOR pathway activity was also analyzed. Finally, the effect of BCH (LAT inhibitor) combined with radiotherapy on GBM prognosis was verified in vivo. RESULTS We have first confirmed that TMZ not only induces resistance to chemotherapy in GBM cells but also enhances their resistance to radiotherapy, which is a significant finding in the process of building TMZ-resistant U87MG GBM cell lines. We then performed comprehensive transcriptomic analysis and identified amino acid metabolism as a potential key factor in radiotherapy resistance. Specifically, we confirmed that the upregulation of LAT4 following chemotherapy enhances leucine metabolism within tumors in vitro and in vivo, thereby modulating the mechanistic target of mTOR pathway and leading to radiotherapy resistance. Of note, the application of inhibitors targeting leucine metabolism was shown to restore the sensitivity of these cells to radiotherapy, highlighting a potential therapeutic strategy for overcoming resistance in GBM. CONCLUSIONS Our study links tumor sensitivity to chemotherapy and radiotherapy and highlights the critical role of LAT4 in activating the mTOR pathway and GBM radiotherapy resistance. It suggests ways to improve radiotherapy sensitivity to GBM.
Collapse
Affiliation(s)
- Wenrui Zang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Yangwu Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Jiajun Zheng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Yifeng Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Lei Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Chiyang Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Jiakun Zhao
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Qiang Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yangheng Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Zhenyuan Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Yongfu Cao
- Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wanling Zhang
- Department of Psychiatry, Guangzhou Tianhe District People's Hospital, Guangzhou, China
| | - Junjie Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China.
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yuntao Lu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, 838 North Guangzhou Ave, Guangzhou, 510515, China.
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Wang B, Yu RZ, Zhang XY, Ren Y, Zhen YW, Han L. Polo-like kinase 4 accelerates glioma malignant progression and vasculogenic mimicry by phosphorylating EphA2. Cancer Lett 2024; 611:217397. [PMID: 39694224 DOI: 10.1016/j.canlet.2024.217397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Vasculogenic mimicry (VM), which involved the formation of vascular-like structures by highly invasive tumor cells, had been identified as one of the mechanisms contributing to resistance against anti-angiogenic therapy in patients with glioblastoma (GBM). Therefore, inhibition of VM formation may serve as an effective therapeutic strategy against angiogenesis resistance. Polo-like kinase 4 (PLK4), a protein kinase, had been linked to the progression of glioblastoma and was associated with an unfavorable prognosis. The integration of proteomics and phosphoproteomics revealed that PLK4 directly activated the PI3K-Akt and MAPK signaling cascades by phosphorylating the Ser901 and Ser897 of EphA2. In addition, EphA2 Ser901 phosphorylating catalyzed by PLK4 significantly enhanced the phosphorylation of its own Ser897 site, which is a hallmark of EphA2 activation. The PI3K-Akt signaling was intricately associated with the progression of VM. Thus, PLK4 influenced malignant progression and VM formation via stimulation of the EphA2 signal transduction. Moreover, the expression level of PLK4 protein positively correlated with the level of EphA2 phosphorylation in glioma tissues. These results highlighted the crucial significance of PLK4 phosphorylating EphA2 in the malignant progression and VM formation in GBM.
Collapse
Affiliation(s)
- Bo Wang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Run-Ze Yu
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiao-Yang Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yu Ren
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Ying-Wei Zhen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Lei Han
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
8
|
Pathikonda S, Amirmahani F, Mathew D, Muthukrishnan SD. Histone acetyltransferases as promising therapeutic targets in glioblastoma resistance. Cancer Lett 2024; 604:217269. [PMID: 39326554 DOI: 10.1016/j.canlet.2024.217269] [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: 06/17/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Glioblastoma (GBM) is a fatal adult brain tumor with an extremely poor prognosis. GBM poses significant challenges for targeted therapies due to its intra- and inter-tumoral heterogeneity, a highly immunosuppressive microenvironment, diffuse infiltration into normal brain parenchyma, protection by the blood-brain barrier and acquisition of therapeutic resistance. Recent studies have implicated epigenetic modifiers as key players driving tumorigenesis, resistance, and progression of GBM. While the vast majority of GBM research on epigenetic modifiers thus far has focused predominantly on elucidating the functional roles and targeting of DNA methyltransferases and histone deacetylases, emerging evidence indicates that histone acetyltransferases (HATs) also play a key role in mediating plasticity and therapeutic resistance in GBM. Here, we will provide an overview of HATs, their dual roles and functions in cancer as both tumor suppressors and oncogenes and focus specifically on their implications in GBM resistance. We also discuss the technical challenges in developing selective HAT inhibitors and highlight their promise as potential anti-cancer therapeutics for treating intractable cancers such as GBM.
Collapse
Affiliation(s)
- Spoorthy Pathikonda
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| | - Farzaneh Amirmahani
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| | - Diya Mathew
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| | - Sree Deepthi Muthukrishnan
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| |
Collapse
|
9
|
Koeffler HP. NBS1 lactylation and damaged DNA repair. Cancer Lett 2024; 604:217128. [PMID: 39179095 DOI: 10.1016/j.canlet.2024.217128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 08/26/2024]
Affiliation(s)
- H Phillip Koeffler
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, USA.
| |
Collapse
|
10
|
Gao Z, Luan X, Wang X, Han T, Li X, Li Z, Li P, Zhou Z. DNA damage response-related ncRNAs as regulators of therapy resistance in cancer. Front Pharmacol 2024; 15:1390300. [PMID: 39253383 PMCID: PMC11381396 DOI: 10.3389/fphar.2024.1390300] [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: 02/23/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
The DNA damage repair (DDR) pathway is a complex signaling cascade that can sense DNA damage and trigger cellular responses to DNA damage to maintain genome stability and integrity. A typical hallmark of cancer is genomic instability or nonintegrity, which is closely related to the accumulation of DNA damage within cancer cells. The treatment principles of radiotherapy and chemotherapy for cancer are based on their cytotoxic effects on DNA damage, which are accompanied by severe and unnecessary side effects on normal tissues, including dysregulation of the DDR and induced therapeutic tolerance. As a driving factor for oncogenes or tumor suppressor genes, noncoding RNA (ncRNA) have been shown to play an important role in cancer cell resistance to radiotherapy and chemotherapy. Recently, it has been found that ncRNA can regulate tumor treatment tolerance by altering the DDR induced by radiotherapy or chemotherapy in cancer cells, indicating that ncRNA are potential regulatory factors targeting the DDR to reverse tumor treatment tolerance. This review provides an overview of the basic information and functions of the DDR and ncRNAs in the tolerance or sensitivity of tumors to chemotherapy and radiation therapy. We focused on the impact of ncRNA (mainly microRNA [miRNA], long noncoding RNA [lncRNA], and circular RNA [circRNA]) on cancer treatment by regulating the DDR and the underlying molecular mechanisms of their effects. These findings provide a theoretical basis and new insights for tumor-targeted therapy and the development of novel drugs targeting the DDR or ncRNAs.
Collapse
Affiliation(s)
- Ziru Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xinchi Luan
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xuezhe Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Tianyue Han
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xiaoyuan Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zeyang Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zhixia Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Zhan Y, Zhang Z, Yin A, Su X, Tang N, Chen Y, Zhang Z, Chen W, Wang J, Wang W. RBBP4: A novel diagnostic and prognostic biomarker for non-small-cell lung cancer correlated with autophagic cell death. Cancer Med 2024; 13:e70090. [PMID: 39109577 PMCID: PMC11304277 DOI: 10.1002/cam4.70090] [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: 11/15/2023] [Revised: 06/07/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) often presents at later stages, typically associated with poor prognosis. Autophagy genes play a role in the progression of tumors. This study investigated the clinical relevance, prognostic value, and biological significance of RBBP4 in NSCLC. METHODS We assessed RBBP4 expression using the GSE30219 and TCGA NSCLC datasets and NSCLC cells, exploring its links with clinical outcomes, tumor immunity, and autophagy genes through bioinformatics analysis after transcriptome sequencing of RBBP4-knockdown and control PC9 cells. We identified differentially expressed genes (DEGs) and conducted Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway enrichment, and protein-protein interaction network analyses. The significance of autophagy-related DEGs was evaluated for diagnosis and prognosis using the GSE30219 dataset. Experiments both in vivo and in vitro explored the biological mechanisms behind RBBP4-mediated autophagic cell death in NSCLC. RESULTS RBBP4 overexpression in NSCLC correlates with a poorer prognosis. Eighteen types of immune cell were significantly enriched in cultures that had low RBBP4 expression compared high expression. DEGs associated with RBBP4 are enriched in autophagy pathways. Transcriptomic profiling of the PC9 cell line identified autophagy-related DEGs associated with RBBP4 that exhibited differential expression in NSCLC, suggesting prognostic applications. In vitro experiments demonstrated that RBBP4 knockdown induced autophagy and apoptosis in PC9 cells, promoting cell death, which was inhibited by 3-MA. In vivo, targeted siRNA against RBBP4 significantly reduced tumor development in PC9 cell-injected nude mice, elevating autophagy-related protein levels and inducing apoptosis and necrosis in tumor tissues. CONCLUSION In NSCLC, RBBP4 upregulation correlates with poor prognosis and altered immunity. Its knockdown induces autophagic cell death in NSCLC cells. These results indicate RBBP4 as a potential NSCLC diagnostic marker and its autophagy modulation as a prospective therapeutic target.
Collapse
Affiliation(s)
- Yajing Zhan
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zhiqian Zhang
- Department of Clinical Laboratory CenterShaoxing People's Hospital (Shaoxing Hospital)ShaoxingZhejiangChina
| | - Ankang Yin
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Xiyang Su
- Department of Laboratory MedicineThe Second Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Nan Tang
- Department of Clinical LaboratoryPeople's Hospital of Wangcheng District ChangshaChangshaHunanChina
| | - Yi Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zebin Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Wei Chen
- Institute of Clinical Medicine Research, Zhejiang Provincial People's Hospital, Hangzhou Medical CollegeHangzhouZhejiangChina
- Cancer Institute of Integrated Tradition Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese MedicineTongde Hospital of Zhejiang ProvinceHangzhouZhejiangChina
| | - Juan Wang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese MedicineTongde Hospital of Zhejiang ProvinceHangzhouZhejiangChina
| | - Wei Wang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese MedicineTongde Hospital of Zhejiang ProvinceHangzhouZhejiangChina
| |
Collapse
|
12
|
Yang E, Hong B, Wang Y, Wang Q, Zhao J, Cui X, Wu Y, Yang S, Su D, Liu X, Kang C. EPIC-0628 abrogates HOTAIR/EZH2 interaction and enhances the temozolomide efficacy via promoting ATF3 expression and inhibiting DNA damage repair in glioblastoma. Cancer Lett 2024; 588:216812. [PMID: 38490327 DOI: 10.1016/j.canlet.2024.216812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/17/2024]
Abstract
The efficacy of temozolomide (TMZ) treatment in glioblastoma (GBM) is influenced by various mechanisms, mainly including the level of O6-methylguanine-DNA methyltransferase (MGMT) and the activity of DNA damage repair (DDR) pathways. In our previous study, we had proved that long non-coding RNA HOTAIR regulated the GBM progression and mediated DDR by interacting with EZH2, the catalytic subunit of PRC2. In this study, we developed a small-molecule inhibitor called EPIC-0628 that selectively disrupted the HOTAIR-EZH2 interaction and promoted ATF3 expression. The upregulation of ATF3 inhibited the recruitment of p300, p-p65, p-Stat3 and SP1 to the MGMT promoter. Hence, EPIC-0628 silenced MGMT expression. Besides, EPIC-0628 induced cell cycle arrest by increasing the expression of CDKN1A and impaired DNA double-strand break repair via suppressing the ATF3-p38-E2F1 pathway. Lastly, EPIC-0628 enhanced TMZ efficacy in GBM in vitro and vivo. Hence, this study provided evidence for the combination of epigenetic drugs EPIC-0628 with TMZ for GBM treatment through the above mechanisms.
Collapse
Affiliation(s)
- Eryan Yang
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China; Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Biao Hong
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yunfei Wang
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qixue Wang
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jixing Zhao
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaoteng Cui
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ye Wu
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Shixue Yang
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Dongyuan Su
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaomin Liu
- Neuro-Oncology Center, Tianjin Huanhu Hospital, Nankai University, Tianjin, 300350, China
| | - Chunsheng Kang
- Lab of Neuro- Oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
13
|
Cai L, Liu B, Cao Y, Sun T, Li Y. Unveiling the molecular structure and role of RBBP4/7: implications for epigenetic regulation and cancer research. Front Mol Biosci 2023; 10:1276612. [PMID: 38028543 PMCID: PMC10679446 DOI: 10.3389/fmolb.2023.1276612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Retinoblastoma-binding protein (RBBP) family is a class of proteins that can interact with tumor suppressor retinoblastoma protein (pRb). RBBP4 and RBBP7 are the only pair of homologous proteins in this family, serving as scaffold proteins whose main function is to offer a platform to indirectly connect two proteins. This characteristic allows them to extensively participate in the binding of various proteins and epigenetic complexes, indirectly influencing the function of effector proteins. As a result, they are often highlighted in organism activities involving active epigenetic modifications, such as embryonic development and cancer activation. In this review, we summarize the structural characteristics of RBBP4/7, the complexes they are involved in, their roles in embryonic development and cancer, as well as potential future research directions, which we hope to inspire the field of epigenetic research in the future.
Collapse
Affiliation(s)
- Lize Cai
- The First Affiliated Hospital of Soochow University, Suzhou University, Suzhou, China
| | - Bin Liu
- Department of Neurosurgery, Qinghai Provincial People’s Hospital, Xining, China
| | - Yufei Cao
- The First Affiliated Hospital of Soochow University, Suzhou University, Suzhou, China
| | - Ting Sun
- The First Affiliated Hospital of Soochow University, Suzhou University, Suzhou, China
| | - Yanyan Li
- The First Affiliated Hospital of Soochow University, Suzhou University, Suzhou, China
| |
Collapse
|
14
|
Chen H, Zhang W, Luo S, Li Y, Zhu Q, Xia Y, Tan H, Bian Y, Li Y, Ma J, Chen W, Luo X, Zhu G. Lead exposure induces neuronal apoptosis via NFκB p65/RBBP4/Survivin signaling pathway. Toxicology 2023; 499:153654. [PMID: 37866543 DOI: 10.1016/j.tox.2023.153654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/08/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Lead (Pb), as a heavy metal that is easily exposed in daily life, can cause damage to various systems of body. Apoptosis is an autonomous cell death process regulated by genes in order to maintain the stability of internal environment, which plays an important role in the development of nervous system. RB binding protein 4 (RBBP4) is one of the core histone binding subunits and is closely related to the apoptosis process of nervous system cells. However, it is not known whether RBBP4 can regulate neuronal apoptosis in lead-exposed environments. We exposed PC12 cells to 0 μM (control group), 1 μM, and 100 μM PbAc for 24 h to obtain cell samples. The female rats ingested drinking water containing 0, 0.5 g/L, and 2.0 g/L PbAc from the first day of pregnancy to three weeks after delivery to obtain hippocampal tissue samples from mammary rats. The results of TUNEL showed that lead exposure promoted the onset of apoptosis in cells and hippocampus. The mRNA and protein levels of the apoptosis-related protein Survivin were significantly reduced in the lead-exposed group compared to the control group. In addition, we found that lead exposure reduces the mRNA and protein levels of RBBP4 in PC12 cells and hippocampus, and increases the mRNA and protein levels of NFκB p65. Moreover, inhibiting NFκB p65 can reverse the decrease in RBBP4 expression in the lead exposure model. Overexpression of RBBP4 increased Survivin expression and reduced apoptosis induced by lead exposure. This suggests that lead exposure induces apoptosis through the NFκB p65/RBBP4/Survivin signaling pathway.
Collapse
Affiliation(s)
- Hui Chen
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Wei Zhang
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Song Luo
- Department of Orthopaedic Surgery, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Diezihu Avenue Honggutan District, Nanchang 330006, PR China
| | - Yanshu Li
- Jiangxi Center of Medical Device Testing, Nanchang 330029, PR China
| | - Qian Zhu
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Yongli Xia
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Hong Tan
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Ying Bian
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Yaobing Li
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Jianmin Ma
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Wei Chen
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Xietian Luo
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Gaochun Zhu
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China.
| |
Collapse
|
15
|
Zhuang X, Xiao R, Fu Y, Yang B, Fan J, Lu F, Qin T, Yang X, Hu X, Yin J, Li W, Kang X, Chen G, Hu D, Sun C. MRE11:p.K464R mutation mediates olaparib resistance by enhancing DNA damage repair in HGSOC. Cell Biosci 2023; 13:178. [PMID: 37759323 PMCID: PMC10537967 DOI: 10.1186/s13578-023-01117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Although the clinical application of PARP inhibitors has brought hope to ovarian cancer, the problem of its resistance has become increasingly prominent. Therefore, clinical experts have been focused on finding specific indicators and therapeutic targets that can be used for resistance monitoring of PARP inhibitors. RESULTS By cfDNA detecting during Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer, we found the presence of MRE11:p.K464R mutation was strongly associated with acquired Olaparib resistance. Structural analysis revealed that the MRE11:p.K464R mutation is situated at a critical site where the MRE11 protein interacts with other biomolecules, leading to potential structural and functional abnormalities of MRE11 protein. Functionally, MRE11:p.K464R mutation enhanced the tolerance of Olaparib by reducing the DNA damage. Mechanistically, MRE11:p.K464R mutation improved the efficiency of DNA damage repair and induce Olaparib resistance by enhancing its binding activity with the interacting proteins (including RAD50 and RPS3). Among them, the enhanced binding of MRE11:p.K464R mutation to RAD50/RPS3 facilitated non-homologous end joining (NHEJ) repair in tumor cells, thereby expanding the scope of research into acquired resistance to PARP inhibitors. CONCLUSIONS Our findings provide a theoretical basis for MRE11:p.K464R mutation as a specific indicator of resistance monitoring in Olaparib treatment, and the exploration of its resistance mechanism provides a novel insights for the formulation of combination ther therapies after Olaparib resistance.
Collapse
Affiliation(s)
- Xucui Zhuang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rourou Xiao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Fu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Yang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junpeng Fan
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Funian Lu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Qin
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohang Yang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingyuan Hu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Yin
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenting Li
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Kang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Chen
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dianxing Hu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chaoyang Sun
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
16
|
Li Y, Gao Z, Wang Y, Pang B, Zhang B, Hu R, Wang Y, Liu C, Zhang X, Yang J, Mei M, Wang Y, Zhou X, Li M, Ren Y. Lysine methylation promotes NFAT5 activation and determines temozolomide efficacy in glioblastoma. Nat Commun 2023; 14:4062. [PMID: 37429858 DOI: 10.1038/s41467-023-39845-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 06/30/2023] [Indexed: 07/12/2023] Open
Abstract
Temozolomide (TMZ) therapy offers minimal clinical benefits in patients with glioblastoma multiforme (GBM) with high EGFR activity, underscoring the need for effective combination therapy. Here, we show that tonicity-responsive enhancer binding protein (NFAT5) lysine methylation, is a determinant of TMZ response. Mechanistically, EGFR activation induces phosphorylated EZH2 (Ser21) binding and triggers NFAT5 methylation at K668. Methylation prevents NFAT5 cytoplasm interaction with E3 ligase TRAF6, thus blocks NFAT5 lysosomal degradation and cytosol localization restriction, which was mediated by TRAF6 induced K63-linked ubiquitination, resulting in NFAT5 protein stabilization, nuclear accumulation and activation. Methylated NFAT5 leads to the upregulation of MGMT, a transcriptional target of NFAT5, which is responsible for unfavorable TMZ response. Inhibition of NFAT5 K668 methylation improved TMZ efficacy in orthotopic xenografts and patient-derived xenografts (PDX) models. Notably, NFAT5 K668 methylation levels are elevated in TMZ-refractory specimens and confer poor prognosis. Our findings suggest targeting NFAT5 methylation is a promising therapeutic strategy to improve TMZ response in tumors with EGFR activation.
Collapse
Affiliation(s)
- Yatian Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhenyue Gao
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuhong Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Bo Pang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Binbin Zhang
- Department of Neuro-oncology, Tianjin Huanhu Hospital, Tianjin, China
| | - Ruxin Hu
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuqing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chao Liu
- Department of Maxillofacial and Otorhinolaryngology Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Xuebin Zhang
- Department of Pathology, Tianjin Huanhu Hospital, Tianjin, China
| | - Jingxuan Yang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mei Mei
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Yongzhi Wang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xuan Zhou
- Department of Maxillofacial and Otorhinolaryngology Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China.
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Yu Ren
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
17
|
Liu S, Zhang HL, Li J, Ye ZP, Du T, Li LC, Guo YQ, Yang D, Li ZL, Cao JH, Hu BX, Chen YH, Feng GK, Li ZM, Deng R, Huang JJ, Zhu XF. Tubastatin A potently inhibits GPX4 activity to potentiate cancer radiotherapy through boosting ferroptosis. Redox Biol 2023; 62:102677. [PMID: 36989572 PMCID: PMC10074938 DOI: 10.1016/j.redox.2023.102677] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Ferroptosis, an iron-dependent lipid peroxidation-driven programmed cell death, is closely related to cancer therapy. The development of druggable ferroptosis inducers and their rational application in cancer therapy are critical. Here, we identified Tubastatin A, an HDAC6 inhibitor as a novel druggable ferroptosis inducer through large-scale drug screening. Tubastatin A directly bonded to GPX4 and inhibited GPX4 enzymatic activity through biotin-linked Tubastatin A putdown and LC/MS analysis, which is independent of its inhibition of HDAC6. In addition, our results showed that radiotherapy not only activated Nrf2-mediated GPX4 transcription but also inhibited lysosome-mediated GPX4 degradation, subsequently inducing ferroptosis tolerance and radioresistance in cancer cells. Tubastatin A overcame ferroptosis resistance and radioresistance of cancer cells by inhibiting GPX4 enzymatic activity. More importantly, Tubastatin A has excellent bioavailability, as demonstrated by its ability to significantly promote radiotherapy-induced lipid peroxidation and tumour suppression in a mouse xenograft model. Our findings identify a novel druggable ferroptosis inducer, Tubastatin A, which enhances radiotherapy-mediated antitumor effects. This work provides a compelling rationale for the clinical evaluation of Tubastatin A, especially in combination with radiotherapy.
Collapse
|