1
|
Liang Y, Du M, Li X, Gao J, Li Q, Li H, Li J, Gao X, Cong H, Huang Y, Li X, Wang L, Cui J, Gan Y, Tu H. Upregulation of Lactobacillus spp. in gut microbiota as a novel mechanism for environmental eustress-induced anti-pancreatic cancer effects. Gut Microbes 2025; 17:2470372. [PMID: 39988618 PMCID: PMC11853549 DOI: 10.1080/19490976.2025.2470372] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with limited effective treatment options. Emerging evidence links enriched environment (EE)-induced eustress to PDAC inhibition. However, the underlying mechanisms remain unclear. In this study, we explored the role of gut microbiota in PDAC-suppressive effects of EE. We demonstrated that depletion of gut microbiota with antibiotics abolished EE-induced tumor suppression, while fecal microbiota transplantation (FMT) from EE mice significantly inhibited tumor growth in both subcutaneous and orthotopic PDAC models housed in standard environment. 16S rRNA sequencing revealed that EE enhanced gut microbiota diversity and selectively enriched probiotic Lactobacillus, particularly L. reuteri. Treatment with L. reuteri significantly suppressed PDAC tumor growth and increased natural killer (NK) cell infiltration into the tumor microenvironment. Depletion of NK cells alleviated the anti-tumor effects of L. reuteri, underscoring the essential role of NK cell-mediated immunity in anti-tumor response. Clinical analysis of PDAC patients showed that higher fecal Lactobacillus abundance correlated with improved progression-free and overall survival, further supporting the therapeutic potential of L. reuteri in PDAC. Overall, this study identifies gut microbiota as a systemic regulator of PDAC under psychological stress. Supplementation of psychobiotic Lactobacillus may offer a novel therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Yiyi Liang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Du
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Gao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huimin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Gao
- School of Basic Medicine, Fudan University, Shanghai, China
| | - Hui Cong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimeng Huang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinran Li
- School of Basic Medicine, Fudan University, Shanghai, China
| | - Liwei Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiujie Cui
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Gan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Tu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Xie Y, Mi X, Xing Y, Dai Z, Pu Q. Past, present, and future of exosomes research in cancer: A bibliometric and visualization analysis. Hum Vaccin Immunother 2025; 21:2488551. [PMID: 40207548 PMCID: PMC11988232 DOI: 10.1080/21645515.2025.2488551] [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: 01/21/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer seriously threatens the lives and health of people worldwide, and exosomes seem to play an important role in managing cancer effectively, which has attracted extensive attention from researchers in recent years. This study aimed to scientifically visualize exosomes research in cancer (ERC) through bibliometric analysis, reviewing the past, summarizing the present, and predicting the future, with a view to providing valuable insights for scholars and policy makers. Researches search and data collection from Web of Science Core Collection and clinical trial.gov. Calculations and visualizations were performed using Microsoft Excel, VOSviewer, Bibliometrix R-package, and CiteSpace. As of December 1, 2024, and March 8, 2025, we identified 8,001 ERC-related publications and 107 ERC-related clinical trials, with an increasing trend in annual publications. Our findings supported that China, Nanjing Medical University, and International Journal of Molecular Sciences were the most productive countries, institutions, and journals, respectively. Whiteside, Theresa L. had the most publications, while Théry, C was the most co-cited scholar. In addition, Cancer Research was the most co-cited journal. Spatial and temporal distribution of clinical trials was the same as for publications. High-frequency keywords were "extracellular vesicle," "microRNA" and "biomarker." Additional, "surface functionalization," "plant," "machine learning," "nanomaterials," "promotes metastasis," "engineered exosomes," and "macrophage-derived exosomes" were promising research topics. Our study comprehensively and visually summarized the structure, hotspots, and evolutionary trends of ERC. It would inspire subsequent studies from a macroscopic perspective and provide a basis for rational allocation of resources and identification of collaborations among researchers.
Collapse
Affiliation(s)
- Yafei Xie
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xingqi Mi
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yikai Xing
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhangyi Dai
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Pu
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Xiao Y, He M, Zhang X, Yang M, Yuan Z, Yao S, Qin Y. Research progress on the mechanism of tumor cell ferroptosis regulation by epigenetics. Epigenetics 2025; 20:2500949. [PMID: 40327848 PMCID: PMC12064064 DOI: 10.1080/15592294.2025.2500949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer remains a significant barrier to human longevity and a leading cause of mortality worldwide. Despite advancements in cancer therapies, challenges such as cellular toxicity and drug resistance to chemotherapy persist. Regulated cell death (RCD), once regarded as a passive process, is now recognized as a programmed mechanism with distinct biochemical and morphological characteristics, thereby presenting new therapeutic opportunities. Ferroptosis, a novel form of RCD characterized by iron-dependent lipid peroxidation and unique mitochondrial damage, differs from apoptosis, autophagy, and necroptosis. It is driven by reactive oxygen species (ROS)-induced lipid peroxidation and is implicated in tumorigenesis, anti-tumor immunity, and resistance, particularly in tumors undergoing epithelial-mesenchymal transition. Moreover, ferroptosis is associated with ischemic organ damage, degenerative diseases, and aging, regulated by various cellular metabolic processes, including redox balance, iron metabolism, and amino acid, lipid, and glucose metabolism. This review focuses on the role of epigenetic factors in tumor ferroptosis, exploring their mechanisms and potential applications in cancer therapy. It synthesizes current knowledge to provide a comprehensive understanding of epigenetic regulation in tumor cell ferroptosis, offering insights for future research and clinical applications.
Collapse
Affiliation(s)
- Yuyang Xiao
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mengyang He
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xupeng Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Meng Yang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhangchi Yuan
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shanhu Yao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Medical Information Research, Central South University, Changsha, Hunan, China
| | - Yuexiang Qin
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Zhong L, Zhu J, Chen J, Jin X, Liu L, Ji S, Luo J, Wang H. MGAT4EP promotes tumor progression and serves as a prognostic marker for breast cancer. Cancer Biol Ther 2025; 26:2475604. [PMID: 40069131 PMCID: PMC11901376 DOI: 10.1080/15384047.2025.2475604] [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: 12/07/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Breast cancer remains a global health challenge with varied prognoses despite treatment advancements. Therefore, this study explores the pseudogene MGAT4EP as a potential biomarker and therapeutic target in breast cancer. Using TCGA data and bioinformatics, MGAT4EP was identified as significantly overexpressed in breast cancer tissues and associated with poor prognosis. Multivariate Cox regression confirmed MGAT4EP as important prognostic factor. A clinical prediction model based on MGAT4EP expression showed high accuracy for 1-, 3-, and 5-year survival rates and was translated into a nomogram for clinical application. Functional studies revealed that silencing MGAT4EP via siRNA promoted apoptosis, inhibited migration and invasion in breast cancer cells. RNA-seq, GSEA, and GO analyses linked MGAT4EP to apoptosis and focal adhesion pathways. Notably, knock down of MGAT4EP significantly suppressed tumor growth and metastasis in xenograft and lung metastasis models. Taken together, these findings establish MGAT4EP as an attractive target for metastatic breast cancer and provide a potential a promising therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Lin Zhong
- Department of Breast Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jianfeng Zhu
- Department of Breast Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jie Chen
- Department of Breast Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xuchu Jin
- Department of Breast Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Liangquan Liu
- Department of Breast Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shufeng Ji
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Luo
- Department of Breast Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hong Wang
- School of Pharmacy, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Fuenteslópez CV, Papapavlou M, Thompson MS, Ye H. Engineering a long-lasting microvasculature in vitro model for traumatic injury research. BIOMATERIALS ADVANCES 2025; 174:214310. [PMID: 40220460 DOI: 10.1016/j.bioadv.2025.214310] [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: 07/31/2024] [Revised: 02/21/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
Microvascular injuries can have systemic physiological effects that exacerbate other injuries and pose a danger to life. Reliable in vitro microvascular models are required to enhance understanding of traumatic injuries. This research aims to develop and optimise a three-dimensional (3D) hydrogel construct for the formation and long-term stability of an in vitro microvascular model for trauma research. First, we develop a 3D hydrogel scaffold using a physiologically relevant cell type to enable the formation of a durable microvascular endothelial network and validate it against the gold standard: HUVECs. Then, we explore the impact of modifying the hydrogel composition, specifically fibrinogen source and concentration, medium, and crosslinking ratio, on scaffold material properties and, consequently, the formation of endothelial networks, their architecture, and long-term integrity. Our results demonstrate that 3D hydrogel scaffolds are crucial for maintaining network stability beyond the initial 24 h. For trauma research applications, the material properties and mechanical behaviour of the hydrogels are critical. Microrheometry revealed that fibrinogen concentration significantly influences gelation times, absorbance rate, storage modulus (G'), loss modulus (G"), and complex viscosity, while also reducing creep compliance. Our multi-pronged approach to engineering microvasculature constructs revealed that variations in hydrogel composition, including fibrinogen concentration and source, crosslinking ratio and choice of medium, strongly affect the hydrogel material characteristics and, in turn, the resulting microvascular networks. Hydrogels made with high concentrations of human fibrinogen, a 200:10:1 crosslinking ratio, and endothelial basal medium (EBM) or EBM supplemented with VEGF performed best, demonstrating superior long-term network stability. The microvasculature construct developed here could be used as a potential platform for studying traumatic injuries, as well as testing interventions aimed at improving recovery and mitigating damage.
Collapse
Affiliation(s)
- Carla Verónica Fuenteslópez
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| | - Mariella Papapavlou
- Department of Materials, University of Oxford, Parks Road, Oxford OX1 3PH, United Kingdom
| | - Mark S Thompson
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| |
Collapse
|
6
|
Xi J, Li Y, Lv L, Tang Z, Liu F, Liu J. H 2O 2/O 2 Self-Supplied Nanoplateform for amplifying oxidative stress to Accelerate Photodynamic/Chemodynamic therapy Cycles. J Colloid Interface Sci 2025; 690:137291. [PMID: 40086336 DOI: 10.1016/j.jcis.2025.137291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/18/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Photodynamic (PDT) and chemodynamic therapies (CDT) relying on reactive oxygen species-mediated treatments mainly face various challenges of hypoxia, endogenous hydrogen peroxide (H2O2) deficiency, and glutathione (GSH) overexpression in the tumor microenvironment. Herein, we propose a novel strategy using a core-shell structured nanocomposite, UCNP@mSiO2@5-ALA-CaO2-Cu(UA@CC). The strategy centers on upconverting NPs and then utilizes mesoporous silica loaded with 5-aminolevulinic acid (5-ALA) to maximize the enrichment of protoporphyrin IX (Pph IX), an intra-tumor photosensitizer. Then in the acidic tumor microenvironment (TME), CaO2 in the outer layer reacts with H2O to form O2, H2O2 and Ca2+, and the released H2O2 serves as an auxiliary "fuel" to induce acceleration of the Fenton-like (Cu2+) reaction and inactivation of the antioxidant GSH enzyme, thus enhancing the tumor cells' Catalysis. Furthermore, under the excitation of a 980 nm laser, 5-ALA-mediated PDT and Cu+-based CDT were initiated. Through interconnected processes of Ca2+ overload, self-supply of H2O2/O2, and enhanced GSH depletion, an accelerated cycling strategy for combined PDT/CDT therapy was established, resulting in amplified oxidative stress and anti-tumor capabilities, which was validated in cancer cells and melanoma mouse models.
Collapse
Affiliation(s)
- Jianying Xi
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yong Li
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Longhao Lv
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Zhengshuai Tang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Fangfang Liu
- Weifang University of Science and Technology, Shouguang, Weifang 262700, China; Shandong Engineering Research Center of Green and High-value Marine Fine Chemical, Weifang 262700, China.
| | - Jinliang Liu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
7
|
Bräutigam K, Skok K, Szymonski K, Rift CV, Karamitopoulou E. Tumor immune microenvironment in pancreatic ductal adenocarcinoma revisited - Exploring the "Space". Cancer Lett 2025; 622:217699. [PMID: 40204149 DOI: 10.1016/j.canlet.2025.217699] [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: 12/21/2024] [Revised: 03/24/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most deadly malignancies with a highly immunosuppressive tumor immune microenvironment (TIME) that hinders effective therapy. PDAC is characterized by significant heterogeneity in immune cell composition, spatial distribution and activation states, which impacts tumor progression and treatment response. Tumour-infiltrating lymphocytes (TILs), including CD4+ T-helper cells, CD8+ cytotoxic T-cells and FOXP3+ regulatory T-cells, play a key role in immune regulation, yet PDAC is largely an immunologically "cold" tumour with limited effector T-cell infiltration. The surrounding cellular microenvironment, particularly Cancer Associated Fibroblasts (CAFs) and macrophages, contributes to immune evasion by promoting a fibrotic and desmoplastic barrier that limits TIL infiltration. The prognostic significance of TILs is increasingly recognized, with higher densities correlating with improved survival, whereas regulatory T-cell infiltration and immunosuppressive stromal interactions are associated with poor outcomes. Emerging therapeutic strategies targeting the TIME (e.g., CAFs), immune checkpoint inhibitors, and TIL-based therapies offer the potential to overcome resistance. Future research must focus on optimizing immunotherapy strategies and unravelling the complex stromal-immune interactions to improve clinical translation.
Collapse
Affiliation(s)
- Konstantin Bräutigam
- Institute of Cancer Research, Centre for Evolution and Cancer, London, SM2 5NG, United Kingdom; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland.
| | - Kristijan Skok
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria; Institute of Biomedical Sciences, Medical Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Krzysztof Szymonski
- Department of Pathomorphology, Jagiellonian University Medical College, Krakow, Poland
| | | | - Eva Karamitopoulou
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Liu X, Lv M, Feng B, Gong Y, Min Q, Wang Y, Wu Q, Chen J, Zhao D, Li J, Zhang W, Zhan Q. SQLE amplification accelerates esophageal squamous cell carcinoma tumorigenesis and metastasis through oncometabolite 2,3-oxidosqualene repressing Hippo pathway. Cancer Lett 2025; 621:217528. [PMID: 39924077 DOI: 10.1016/j.canlet.2025.217528] [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: 10/18/2024] [Revised: 01/17/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent cancers worldwide, characterized by a dismal prognosis and elusive therapeutic targets. Dysregulated cholesterol metabolism is a critical hallmark of cancer cells, facilitating tumor progression. Here, we used whole genome sequencing data from several ESCC cohorts to identify the important role of squalene epoxidase (SQLE) in promoting ESCC tumorigenesis and metastasis. Specifically, our findings highlight the significance of 2,3-oxidosqualene, an intermediate metabolite of cholesterol biosynthesis, synthesized by SQLE and metabolized by lanosterol synthase (LSS), as a key regulator of ESCC progression. Mechanistically, the interaction between 2,3-oxidosqualene and vinculin enhances the nuclear accumulation of Yes-associated protein 1 (YAP), thereby increasing YAP/TEAD-dependent gene expression and accelerating both tumor growth and metastasis. In a 4-nitroquinoline 1-oxide (4-NQO)-induced ESCC mouse model, overexpression of Sqle resulted in accelerated tumorigenesis compared to wild-type controls, highlighting the pivotal role of SQLE in vivo. Furthermore, elevated SQLE expression in ESCC patients correlates with a poorer prognoses, suggesting potential therapeutic avenues for treatment. In conclusion, our study elucidates the oncogenic function of 2,3-oxidosqualene as a naturally occurring metabolite and proposes modulation of its levels as a promising therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Xuesong Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Peking University International Cancer Institute, Beijing, 100191, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Mengzhu Lv
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Bicong Feng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Ying Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jie Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Dongyu Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jinting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Weimin Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China.
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Peking University International Cancer Institute, Beijing, 100191, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Soochow University Cancer Institute, Suzhou, 215127, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China.
| |
Collapse
|
9
|
Mikolaskova I, Gidron Y, Durmanova V, Suchankova M, Bucova M, Hunakova L. Mental distress and inflammation in bladder cancer: The nerve makes things less vague. Brain Behav Immun Health 2025; 46:100995. [PMID: 40343109 PMCID: PMC12059323 DOI: 10.1016/j.bbih.2025.100995] [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: 10/23/2024] [Revised: 03/14/2025] [Accepted: 04/12/2025] [Indexed: 05/11/2025] Open
Abstract
Objectives This study aimed to explore the interaction between perceived stress, life satisfaction, heart rate variability (HRV), and immune-inflammatory markers in bladder cancer patients. We investigated how HRV moderates the relationship between psychological distress and levels of TNF-α and TGF-β cytokines. We hypothesized that high vagal nerve activity, as indicated by higher HRV, mitigates the impact of perceived stress and life dissatisfaction on inflammation. Methods The study included 73 patients with bladder cancer. HRV was determined from a 5-min ECG recording, focusing on the standard deviation of normal-to-normal interbeat intervals (SDNN). Psychological distress was measured using the Perceived Stress Scale (PSS), and life satisfaction was evaluated with the Life Satisfaction Questionnaire (LSQ). Serum concentrations of TNF-α and plasma levels of TGF-β were determined using sandwich ELISA. Results We found evidence that HRV modulates the relation between perceived stress and inflammation. In patients with low HRV (SDNN <20 ms), PSS was positively correlated with serum level of TNF-α and negatively with the level of TGF-β, while life satisfaction was positively correlated with TGF-β. These relationships were not significant in patients with high HRV (SDNN ≥20 ms). Conclusion Our findings suggest that high vagal activity, as indicated by higher HRV, may mitigate the adverse effects of psychological distress on immune-inflammatory responses in patients with bladder cancer. Stress-related inflammation took place under conditions of low HRV, highlighting the potential role of autonomic regulation in cancer prognosis. Future research should further explore these relationships to develop interventions aimed at improving patient outcomes through stress management and enhanced vagal nerve activity to regulate inflammation in cancer.
Collapse
Affiliation(s)
- Iveta Mikolaskova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08, Bratislava, Slovakia
| | - Yori Gidron
- Department of Nursing, Faculty of Welfare and Health Sciences, University of Haifa, Haifa, 3498838, Israel
| | - Vladimira Durmanova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08, Bratislava, Slovakia
| | - Magda Suchankova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08, Bratislava, Slovakia
| | - Maria Bucova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08, Bratislava, Slovakia
| | - Luba Hunakova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske namestie 14, 811 08, Bratislava, Slovakia
| |
Collapse
|
10
|
Zhao R, Cao B, Li H, Gao J, Xu Q, Cui H, Yuan Z, Ren H, Wei B. MZT1 protects gastric cancer against glucose starvation through targeting NEDD1. Life Sci 2025; 372:123622. [PMID: 40204068 DOI: 10.1016/j.lfs.2025.123622] [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: 09/12/2024] [Revised: 03/19/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
A fasting mimic diet (FMD) has been proven to be a potential therapeutic regimen for gastric cancer (GC) patients. However, the intolerance of energy restriction and limited efficacies hinder wide application of FMD. To identify critical targets mediating resistance against glucose starvation and explore novel approaches to GC therapy, proteomics profiling was performed to depict the landscape of protein expression changes in cells under glucose deprivation. MZT1 was found to be greatly upregulated. We next investigated potential clinical value and regulatory functions of MZT1. Compared to adjacent normal tissues, MZT1 was upregulated in GC specimens and associated with unfavorable patient prognosis. Both in vitro and in vivo experiments indicated that downregulation of the MZT1 level inhibited GC proliferation, migration, invasion, glycolysis and sensitized cells to glucose starvation. Mechanistically, MZT1 functioned as an oncogenic factor by inhibiting NEDD1 ubiquitination and increasing its expression. In conclusion, during glucose starvation, MZT1 is upregulated in GC cells, which promotes resistance by directly suppression of NEDD1 ubiquitination. Our findings unveil the novel mechanisms by which MZT1 can promote GC malignancy. The potential clinical value of MZT1 as GC biomarkers has been first revealed. Suppression of MZT1 may become a promising approach to improve FMD efficacy, which require further validation by future investigations.
Collapse
Affiliation(s)
- Ruiyang Zhao
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Bo Cao
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Hanghang Li
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Jingwang Gao
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Qixuan Xu
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Hao Cui
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Zhen Yuan
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Huiguang Ren
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Bo Wei
- Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China.
| |
Collapse
|
11
|
Peng X, Feng J, Yang H, Xia P, Pu F. Nrf2: A key regulator in chemoradiotherapy resistance of osteosarcoma. Genes Dis 2025; 12:101335. [PMID: 40242036 PMCID: PMC12000747 DOI: 10.1016/j.gendis.2024.101335] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/24/2024] [Accepted: 04/03/2024] [Indexed: 04/18/2025] Open
Abstract
Osteosarcoma (OS), frequently observed in children and adolescents, is one of the most common primary malignant tumors of the bone known to be associated with a high capacity for invasion and metastasis. The incidence of osteosarcoma in children and adolescents is growing annually, although improvements in survival remain limited. With the clinical application of neoadjuvant chemotherapy, chemotherapy combined with limb-preserving surgery has gained momentum as a major intervention. However, certain patients with OS experience treatment failure owing to chemoradiotherapy resistance or metastasis. Nuclear factor E2-related factor 2 (Nrf2), a key antioxidant factor in organisms, plays a crucial role in maintaining cellular physiological homeostasis; however, its overactivation in cancer cells restricts reactive oxygen species production, promotes DNA repair and drug efflux, and ultimately leads to chemoradiotherapy resistance. Recent studies have also identified the functions of Nrf2 beyond its antioxidative function, including the promotion of proliferation, metastasis, and regulation of metabolism. The current review describes the multiple mechanisms of chemoradiotherapy resistance in OS and the substantial role of Nrf2 in the signaling regulatory network to elucidate the function of Nrf2 in promoting OS chemoradiotherapy resistance and formulating relevant therapeutic strategies.
Collapse
Affiliation(s)
- Xianglin Peng
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| | - Jing Feng
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| | - Han Yang
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Ping Xia
- Department of Orthopedics, Wuhan Fourth Hospital, Wuhan 430030, China
| | - Feifei Pu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| |
Collapse
|
12
|
Zhang Y, Guan X, Chai Y, Lu T, An N, Lin X, Liao X. Rational design, optimization, and biological evaluation of novel pyrrolo-pyridone derivatives as potent and orally active Cbl-b inhibitors. Eur J Med Chem 2025; 290:117488. [PMID: 40120499 DOI: 10.1016/j.ejmech.2025.117488] [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: 01/12/2025] [Revised: 02/23/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b), a member of the Cbl family of RING finger E3 ubiquitin ligases, plays a critical role in negatively regulating T-cell, natural killer (NK) cell, and B-cell activation. Inhibiting Cbl-b has emerged as a promising immuno-oncology strategy to enhance immune cell function. Here, we describe the rational design and optimization of pyrrolo-pyridone derivatives as potent Cbl-b inhibitors. Using structure-based drug design, we identified key structural elements that enhance binding affinity and inhibitory potency. Notably, compound B2 stands out, showing superior potency in stimulating IL-2 production in T cells and modulating phosphorylation of key proteins in T-cell receptor signaling. Furthermore, B2 demonstrates favorable pharmacokinetics and significantly inhibits tumor growth in vivo, outperforming NX-1607, which is currently in clinical trials.
Collapse
Affiliation(s)
- Yixuan Zhang
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, 100084, China; School of Pharmacy, Bengbu Medical University, Bengbu, 233030, China
| | - Xiangna Guan
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yushuang Chai
- Zhuhai Yufan Biotechnologies Co., Ltd, Zhuhai, Guangdong, China
| | - Tingting Lu
- Zhuhai Yufan Biotechnologies Co., Ltd, Zhuhai, Guangdong, China
| | - Na An
- Zhuhai Yufan Biotechnologies Co., Ltd, Zhuhai, Guangdong, China
| | - Xinyu Lin
- Zhuhai Yufan Biotechnologies Co., Ltd, Zhuhai, Guangdong, China.
| | - Xuebin Liao
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
13
|
Clauser P, Pötsch N, Santonocito A, Ferrara F, Zeitouni L, Hörnig M, Weber M, Baltzer PAT, Helbich TH. Contrast-Enhanced Digital Breast Tomosynthesis Compared With Contrast-Enhanced Mammography and Magnetic Resonance Imaging in the Assessment of Breast Lesions: A Pilot Study. Invest Radiol 2025; 60:369-375. [PMID: 39621875 DOI: 10.1097/rli.0000000000001138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
OBJECTIVES Contrast-enhanced mammography (CEM) is an accurate competitor for contrast-enhanced breast magnetic resonance imaging (CE-MRI), but the examination is limited by the lack of 3D information. Digital breast tomosynthesis (DBT) allows better lesion detection and characterization compared with mammography. The availability of quasi-3D contrast imaging could further improve the performance of CEM. The aim of our analysis was to compare the diagnostic performance of a contrast-enhanced digital breast tomosynthesis prototype (CE-DBTp) to CEM and to CE-MRI. MATERIALS AND METHODS This prospective study was approved by the ethics committee, and all patients gave written informed consent. Women who presented with suspicious findings on mammography, DBT, or ultrasound were invited to participate in the study. Participants underwent CEM and CE-DBTp of the breast with the suspicious findings as well as bilateral CE-MRI. Histology was used as the standard of reference. Four readers (R1 and R2 non-experienced; R3 and R4 experienced) evaluated the images, blinded to patients' history, previous imaging, and histology. The readers evaluated CEM, CE-DBTp, and CE-MRI in separate sessions and gave a BI-RADS score for each finding. Sensitivity, specificity, lesion conspicuity, and readers' confidence were calculated and compared. RESULTS We included 84 patients (mean age, 56 years; range, 39-70) with 91 histologically verified breast lesions (27 benign, 64 malignant). The accuracy of the CE-DBTp was high, but significant differences were seen between experienced (both 86.8%) and non-experienced readers (76.9% and 78%, P = 0.021). No differences were found between CEM and CE-DBTp, whereas the accuracy of CE-MRI was higher ( P = 0.002). Sensitivity with CE-DBTp varied (89.1% to 100%) between experienced and non-experienced readers ( P = 0.074), and it was comparable to CEM but lower than CE-MRI ( P = 0.003). Specificity was variable between readers with all modalities. Lesion conspicuity was higher for the CE-DBTp and CE-MRI than for CEM, and confidence was significantly higher with the CE-DBTp than with CEM for one of the readers ( P < 0.001). CONCLUSIONS A high sensitivity and good accuracy were achieved with the CE-DBTp. Lesion conspicuity and readers' confidence were higher with the CE-DBTp compared with CEM. However, CE-MRI had the highest sensitivity and accuracy.
Collapse
Affiliation(s)
- Paola Clauser
- From the Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria (P.C., N.P., A.S., M.W., P.A.T.B., T.H.H.); Catholic University of the Sacred Heart, Institute of Radiology, Rome, Italy (F.F.); Department of Radiology Section of Breast Imaging, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia (L.Z.); and Diagnostic Imaging, Siemens Healthcare GmbH, Forchheim, Germany (M.H.)
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Wang P, Ma Y, Rao X, Luo Q, Xiao X, Wang T, Long F. Kaempferol targets Src to exert its chemopreventive effects on mammary tumorigenesis via regulation of the PI3K/AKT pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156701. [PMID: 40220416 DOI: 10.1016/j.phymed.2025.156701] [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: 09/20/2024] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Breast cancer (BC) is a prevalent malignancy that poses significant risks to the health of women worldwide. The incidence and mortality rates of BC continue to be high, despite improvements in diagnosis and treatment, indicating a need for novel prevention strategies. Kaempferol (KAM) is a common dietary flavonoid with known antitumour properties, but its role in the chemoprevention of BC and the underlying mechanisms largely unexplored. PURPOSE This study aimed to evaluate the chemopreventive effects of KAM on carcinogen-induced BC in vivo and in vitro and to elucidate the underlying molecular mechanisms. METHODS In this study, we used an N-methyl-N-nitrosourea (NMU)-induced rat model of BC and 17β-oestradiol (E2)-treated MCF-10A cells to evaluate the chemopreventive effects of KAM on mammary tumorigenesis. The antioxidant capacity of KAM was assessed by measuring oxidative damage marker levels and antioxidant enzyme expression. Flow cytometry and Hoechst 33258 staining were utilized to analyse cell cycle distribution and apoptosis. The core target of KAM was identified by network pharmacology and validated by molecular docking, MD simulation, CESTA, and BLI. KEGG enrichment analysis, molecular biology tests and the application of specific protein inhibitors were conducted to elucidate the molecular mechanisms modulated by KAM. RESULTS In vivo, KAM inhibited the progression of mammary tumours and delayed pathological changes in the morphological structure of mammary gland cells to varying degrees. In vitro, KAM reduced cell viability, migration, and anchorage-independent growth while triggering cell cycle arrest and apoptosis in E2-treated MCF-10A cells. Furthermore, KAM increased cellular antioxidant capacity and attenuated E2-induced oxidative stress. Mechanistically, KAM directly interacted with Src and inhibited its phosphorylation, thus leading to PI3K/AKT pathway inhibition. Notably, the inhibition of E2-induced cell migration and anchorage-independent growth in vitro by Src- or PI3K/AKT pathway-specific inhibitors was not further enhanced when the cells were cultured with KAM. CONCLUSION In summary, KAM targets the Src-mediated PI3K/AKT pathway to reduce oxidative stress and facilitate apoptosis and cell cycle arrest, thereby inhibiting mammary tumorigenesis. Our study is the first to identify Src kinase as a direct target of KAM in mammary tumorigenesis. These findings give significant perspectives on the potential application of KAM in BC chemoprevention.
Collapse
Affiliation(s)
- Pinghan Wang
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China
| | - Yu Ma
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Xiaohui Rao
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Qianwen Luo
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China
| | - Xiao Xiao
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China
| | - Ting Wang
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China.
| | - Fangyi Long
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China.
| |
Collapse
|
15
|
Moskal K, Khurana N, Siegert L, Lee YS, Clevers H, Elinav E, Puschhof J. Modeling cancer-microbiome interactions in vitro: A guide to co-culture platforms. Int J Cancer 2025; 156:2053-2067. [PMID: 39716471 PMCID: PMC11970552 DOI: 10.1002/ijc.35298] [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: 07/15/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 12/25/2024]
Abstract
The biology of cancer is characterized by an intricate interplay of cells originating not only from the tumor mass, but also its surrounding environment. Different microbial species have been suggested to be enriched in tumors and the impacts of these on tumor phenotypes is subject to intensive investigation. For these efforts, model systems that accurately reflect human-microbe interactions are rapidly gaining importance. Here we present a guide for selecting a suitable in vitro co-culture platform used to model different cancer-microbiome interactions. Our discussion spans a variety of in vitro models, including 2D cultures, tumor spheroids, organoids, and organ-on-a-chip platforms, where we delineate their respective advantages, limitations, and applicability in cancer microbiome research. Particular focus is placed on methodologies that facilitate the exposure of cancer cells to microbes, such as organoid microinjections and co-culture on microfluidic devices. We highlight studies offering critical insights into possible cancer-microbe interactions and underscore the importance of in vitro models in those discoveries. We anticipate the integration of more complex microbial communities and the inclusion of immune cells into co-culture systems to more accurately simulate the tumor microenvironment. The advent of ever more sophisticated co-culture models will aid in unraveling the mechanisms of cancer-microbiome interplay and contribute to exploiting their potential in novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Kamil Moskal
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- DKFZ Hector Cancer Institute at the University Medical CenterMannheimGermany
| | - Nimisha Khurana
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Luisa Siegert
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
| | - Ye Seul Lee
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Hans Clevers
- Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC UtrechtHubrecht InstituteUtrechtThe Netherlands
- Present address:
Roche Pharmaceutical Research and Early DevelopmentBaselSwitzerland
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Jens Puschhof
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- DKFZ Hector Cancer Institute at the University Medical CenterMannheimGermany
| |
Collapse
|
16
|
Nordengen AL, Krutto A, Kværner AS, Alavi DT, Henriksen HB, Kolle Å, Henriksen C, Smeland S, Bøhn SK, Zheng C, Shaposhnikov S, Collins AR, Blomhoff R. Plant-based diet and oxidative stress-induced DNA damage in post-surgery colorectal cancer patients: Results from a randomized controlled trial. Free Radic Biol Med 2025; 233:240-249. [PMID: 40180023 DOI: 10.1016/j.freeradbiomed.2025.03.047] [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: 02/10/2025] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
Oxidative stress-induced DNA damage may impact long-term outcomes in colorectal cancer (CRC) patients. While bioactive compounds in plant foods have been linked to DNA protection, evidence among patients in remission remains limited. The present study aimed to investigate the effect of a one-year personalized intensive dietary intervention on DNA damage in post-surgery, non-metastatic CRC patients. Participants were enrolled 2-9 months after surgery in the ongoing randomized controlled trial, Norwegian dietary guidelines and colorectal cancer survival (CRC-NORDIET). Eligible participants (aged 50-80 years, primary stage I-III CRC) were randomized to either a plant-based dietary intervention targeting oxidative stress and inflammation, or to a control group that received standard dietary advice as a part of routine cancer care. As a secondary analysis, this study included 156 participants (78 in the intervention group and 78 in the control group) from the total 503 patients enrolled in CRC-NORDIET study. DNA damage in peripheral mononuclear blood cells (PBMCs) was assessed using the enzyme-modified comet assay during a 12-month follow-up period. A significant intervention effect on DNA base oxidation from baseline to 12 months was observed (P = 0.04), representing a 32 % reduction in the intervention group compared to the control group. No significant effect on DNA strand breaks was found. In conclusion, adherence to a plant-based dietary pattern may reduce DNA base oxidation in post-surgery CRC patients. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT01570010.
Collapse
Affiliation(s)
- Anne Lene Nordengen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway; Norgenotech AS, Oslo Cancer Cluster Incubator, Oslo, Norway; Department of Sport Science and Physical Education, Faculty of Health and Sport Sciences, University of Agder, Kristiansand, Norway.
| | - Annika Krutto
- Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Ane S Kværner
- Section for Colorectal Cancer Screening, The Cancer Registry of Norway, Oslo, Norway
| | - Dena T Alavi
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Hege B Henriksen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Åshild Kolle
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Christine Henriksen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Sigbjørn Smeland
- Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Norway, Oslo, Norway
| | - Siv K Bøhn
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Congying Zheng
- Norgenotech AS, Oslo Cancer Cluster Incubator, Oslo, Norway; Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translation Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | | | - Andrew R Collins
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway; Norgenotech AS, Oslo Cancer Cluster Incubator, Oslo, Norway
| | - Rune Blomhoff
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway; Department of Clinical Service, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
17
|
Oh JM, Guo T, Begum HM, Marty SE, Sha L, Kilic C, Zhou H, Dou Y, Shen K. A micro-metabolic rewiring assay for assessing hypoxia-associated cancer metabolic heterogeneity. Bioact Mater 2025; 48:493-509. [PMID: 40093303 PMCID: PMC11910375 DOI: 10.1016/j.bioactmat.2025.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/11/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Cancer metabolism plays an essential role in therapeutic resistance, where significant inter- and intra-tumoral heterogeneity exists. Hypoxia is a prominent driver of metabolic rewiring behaviors and drug responses. Recapitulating the hypoxic landscape in the tumor microenvironment thus offers unique insights into heterogeneity in metabolic rewiring and therapeutic responses, to inform better treatment strategies. There remains a lack of scalable tools that can readily interface with imaging platforms and resolve the heterogeneous behaviors in hypoxia-associated metabolic rewiring. Here we present a micro-metabolic rewiring (μMeRe) assay that provides the scalability and resolution needed to characterize the metabolic rewiring behaviors of different cancer cells in the context of hypoxic solid tumors. Our assay generates hypoxia through cellular metabolism without external gas controls, enabling the characterization of cell-specific intrinsic ability to drive hypoxia and undergo metabolic rewiring. We further developed quantitative metrics that measure the metabolic plasticity through phenotypes and gene expression. As a proof-of-concept, we evaluated the efficacy of a metabolism-targeting strategy in mitigating hypoxia- and metabolic rewiring-induced chemotherapeutic resistance. Our study and the scalable platform thus lay the foundation for designing more effective cancer treatments tailored toward specific metabolic rewiring behaviors.
Collapse
Affiliation(s)
- Jeong Min Oh
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Tianze Guo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Hydari Masuma Begum
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Saci-Elodie Marty
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Liang Sha
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Cem Kilic
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Hao Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Yali Dou
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
- USC Stem Cell, University of Southern California, Los Angeles, CA, 90033, USA
| |
Collapse
|
18
|
Elkrief A, Routy B, Derosa L, Bolte L, Wargo JA, McQuade JL, Zitvogel L. Gut Microbiota in Immuno-Oncology: A Practical Guide for Medical Oncologists With a Focus on Antibiotics Stewardship. Am Soc Clin Oncol Educ Book 2025; 45:e472902. [PMID: 40262063 DOI: 10.1200/edbk-25-472902] [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: 04/24/2025]
Abstract
The gut microbiota has emerged as a critical determinant of immune checkpoint inhibitor (ICI) efficacy, resistance, and toxicity. Retrospective and prospective studies profiling the taxonomic composition of intestinal microbes of patients treated with ICI have revealed specific gut microbial signatures associated with response. By contrast, dysbiosis, which can be caused by chronic inflammatory processes (such as cancer) or comedications, is a risk factor of resistance to ICI. Recent large-scale meta-analyses have confirmed that antibiotic (ATB) use before or during ICI therapy alters the microbiota repertoire and significantly shortens overall survival, even after adjusting for prognostic factors. These results underscore the importance of implementing ATB stewardship recommendations in routine oncology practice. Microbiota-centered interventions are now being explored to treat gut dysbiosis and optimize ICI responses. Early-phase clinical trials evaluating fecal microbiota transplantation (FMT) from ICI responders or healthy donors have shown that this approach is safe and provided preliminary data on potential efficacy to overcome both primary and secondary resistance to ICI in melanoma, non-small cell lung cancer, and renal cell carcinoma. More targeted interventions including live bacterial products including Clostridium butyricum and Akkermansia massiliensis represent novel microbiome-based adjunct therapies. Likewise, dietary interventions, such as high-fiber diets, have shown promise in enhancing ICI activity. In this ASCO Educational Book, we summarize the current state-of-the-evidence of the clinical relevance of the intestinal microbiota in cancer immunotherapy and provide a practical guide for ATB stewardship.
Collapse
Affiliation(s)
- Arielle Elkrief
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Bertrand Routy
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Lisa Derosa
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
| | - Laura Bolte
- Department of Medical Oncology, University Groningen and University Medical Center, Groningen, the Netherlands
- Department of Gastroenterology and Hepatology, University Groningen and University Medical Center, Groningen, the Netherlands
| | | | | | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| |
Collapse
|
19
|
García-Aranda M, Abitei C, Martín-García D, Fúnez R, Rivas-Ruiz F, Robles-Lasarte M, Alcaide-Garcia J, Téllez T, Aguirre U, Borrero-Martín JJ, Del Cura I, Morales Suárez-Varela MM, Quintana JM, Redondo M. Long-term prognostic value of apoptotic index in colorectal cancer: a 5-year multicentre cohort study. Pathology 2025; 57:443-449. [PMID: 40000339 DOI: 10.1016/j.pathol.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/03/2024] [Accepted: 11/13/2024] [Indexed: 02/27/2025]
Abstract
Despite TNM (Tumour, Node, Metastasis) classification being the gold standard for estimating the prognosis of early-stage colorectal cancer, significant variability in long-term survival persists among patients within the same TNM stage, underscoring the importance of the disease's biological heterogeneity and the need for novel markers. This study investigates the determinants of 5-year mortality in patients with colon or rectal cancer through the analysis of 448 diagnostic tumour samples from a prospective multicentre cohort. We assessed sociodemographic, clinical, and pathological data, as well as the apoptotic index (AI) measured by the terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL) technique. Overall survival was the primary outcome, and Cox regression was used to estimate the hazard ratio (HR). Multivariate 5-year survival analysis identified the highest risk associated with TNM stages IV [p<0.001, HR 12.06, confidence interval (CI) 5.75-25.31] and III (p<0.001, HR 3.52, CI 1.88-6.62), followed by an AI >1.8% (p<0.001, HR 2.16, CI 1.46-3.20), male biological sex (p<0.05, HR 1.58, CI 1.05-2.37), tumour location on the right colon (p<0.024, HR 1.55, CI 1.06-2.27), and age (p<0.001, HR 1.05, CI 1.04-1.07). Our findings underscore the long-term prognostic value of a high AI as a determinant of poor prognosis in colorectal cancer and highlight the need to refine conventional prognostic markers to enable more precise risk stratification.
Collapse
Affiliation(s)
- Marilina García-Aranda
- Research and Innovation Unit, Costa del Sol University Hospital, Marbella, Spain; Department of Surgical Specialties, Biochemistry and Immunology, University of Malaga, Malaga, Spain; Malaga Biomedical Research Institute (IBIMA-BIONAND), Malaga, Spain; Research Network on Chronicity, Primary Care and Prevention and Health Promotion (RICAPPS), Spain.
| | - Cristina Abitei
- Pathology Laboratory, Costa del Sol University Hospital, Marbella, Spain
| | - Desirée Martín-García
- Department of Surgical Specialties, Biochemistry and Immunology, University of Malaga, Malaga, Spain; Malaga Biomedical Research Institute (IBIMA-BIONAND), Malaga, Spain
| | - Rafael Fúnez
- Pathology Laboratory, Costa del Sol University Hospital, Marbella, Spain
| | - Francisco Rivas-Ruiz
- Research and Innovation Unit, Costa del Sol University Hospital, Marbella, Spain; Research Network on Chronicity, Primary Care and Prevention and Health Promotion (RICAPPS), Spain
| | | | | | - Teresa Téllez
- Department of Surgical Specialties, Biochemistry and Immunology, University of Malaga, Malaga, Spain; Malaga Biomedical Research Institute (IBIMA-BIONAND), Malaga, Spain
| | - Urko Aguirre
- Research Network on Chronicity, Primary Care and Prevention and Health Promotion (RICAPPS), Spain; Ikerkuntza Unitatea, Research Unit, Galdakao University Hospital, Galdakao, Spain
| | - Juan José Borrero-Martín
- Department of Hematology, Virgen del Rocío University Hospital, Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Isabel Del Cura
- Research Network on Chronicity, Primary Care and Prevention and Health Promotion (RICAPPS), Spain; Primary Care Research Unit, Primary Care Management, Madrid, Spain; Department of Medical Specialties and Public Health, Rey Juan Carlos University, Madrid, Spain; Research Network on Health Services and Chronic Diseases (REDISSEC), Spain; Gregorio Marañon Health Research Institute (IiSGM), Madrid, Spain
| | - María Manuela Morales Suárez-Varela
- Research Network on Chronicity, Primary Care and Prevention and Health Promotion (RICAPPS), Spain; Department of Preventive Medicine, Unit of Public Health and Environmental Care, University of Valencia, Valencia, Spain
| | - José María Quintana
- Research Network on Chronicity, Primary Care and Prevention and Health Promotion (RICAPPS), Spain; Research Unit Galdakao-Usansolo Hospital, Galdakao, Spain
| | - Maximino Redondo
- Research and Innovation Unit, Costa del Sol University Hospital, Marbella, Spain; Department of Surgical Specialties, Biochemistry and Immunology, University of Malaga, Malaga, Spain; Malaga Biomedical Research Institute (IBIMA-BIONAND), Malaga, Spain; Research Network on Chronicity, Primary Care and Prevention and Health Promotion (RICAPPS), Spain.
| |
Collapse
|
20
|
Zhang D, Zhu Y, Shen Z, Ma S, Liu S, Lu Z. Immunosenescence and immunotherapy in elderly patients with hepatocellular carcinoma. Semin Cancer Biol 2025; 111:60-75. [PMID: 40020977 DOI: 10.1016/j.semcancer.2025.02.010] [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: 12/28/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
Liver cancer, more specifically hepatocellular carcinoma (HCC), is a global health issue and one of the dominant causes of cancer death around the world. In the past few decades, remarkable advances have been achieved in the systemic therapy of HCC. Immune checkpoint inhibitors (ICIs) have become a therapy mainstay for advanced HCC and have shown promise in the neoadjuvant therapy before resection. Despite these significant advancements, the compositions and functions of the immune system occur various alterations with age, called "immunosenescence", which may affect the antitumor effects and safety of ICIs, thus raising concerns that immunosenescence may impair elderly patients' response to ICIs. Therefore, it is important to learn more about the immunosenescence characteristics of elderly patients. However, the real-world elderly HCC patients may be not accurately represented by the elderly patients included in the clinical trials, affecting the generalizability of the efficacy and safety profiles from the clinical trials to the real-world elderly patients. This review summarizes the characteristics of immunosenescence and its influence on HCC progression and immunotherapy efficacy as well as provides the latest progress in ICIs available for HCC and discusses their treatment efficacy and safety on elderly patients. In the future, more studies are needed to clarify the mechanisms of immunosenescence in HCC, and to find sensitive screening tools or biomarkers to identify the patients who may benefit from ICIs.
Collapse
Affiliation(s)
- Dengyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Yan Zhu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhengchao Shen
- Department of General Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, China
| | - Shuoshuo Ma
- Department of General Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Sihua Liu
- Department of General Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Zheng Lu
- Department of General Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China.
| |
Collapse
|
21
|
Wang B, Shi R, Du W, Guo J, He N, Zhu Y, Yu H, Lu H, Zhong L, Li X, Zhou W, Yang F, Feng X. Prodigiosin inhibits proliferation and induces apoptosis through influencing amino acid metabolism in multiple myeloma. Bioorg Chem 2025; 159:108349. [PMID: 40086187 DOI: 10.1016/j.bioorg.2025.108349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
The recurrence of drug-resistant and expensive treatment drugs are major causes of the low survival rate of multiple myeloma (MM) patients. Exploring a safe, effective, low-cost and novel drug treatment for MM is a promising strategy to relieve the burden of MM patients. In this study, we found that prodigiosin could inhibit MM cell proliferation and induce MM cell apoptosis, however, it had a lesser cytotoxic effect on normal B cells within the IC50 range of MM cells. In addition, prodigiosin could inhibit the growth of xenograft MM cells in mice. Transcriptomics and targeted amino acid metabolomics confirmed that prodigiosin could regulate amino acid metabolism, and decrease in amino acid utilization by down-regulated aminoacyl tRNA synthetases expression, resulting in slower growth of MM. In conclusion, prodigiosin exerts anticancer effects on MM cells by interfering with the use of amino acids, indicating its potential novel therapeutic application in MM.
Collapse
Affiliation(s)
- Bingjie Wang
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Furong Laboratory, Xiangya School of Public Health, Central South University, 172# Tongzipo Road, Changsha, Hunan 410013, China
| | - Rui Shi
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Furong Laboratory, Xiangya School of Public Health, Central South University, 172# Tongzipo Road, Changsha, Hunan 410013, China
| | - Wanqing Du
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Furong Laboratory, Xiangya School of Public Health, Central South University, 172# Tongzipo Road, Changsha, Hunan 410013, China
| | - Jiaojiao Guo
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Nihan He
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410078, China
| | - Yinghong Zhu
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410078, China
| | - Han Yu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Furong Laboratory, Xiangya School of Public Health, Central South University, 172# Tongzipo Road, Changsha, Hunan 410013, China
| | - Hongyu Lu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Furong Laboratory, Xiangya School of Public Health, Central South University, 172# Tongzipo Road, Changsha, Hunan 410013, China
| | - Liyuan Zhong
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Furong Laboratory, Xiangya School of Public Health, Central South University, 172# Tongzipo Road, Changsha, Hunan 410013, China
| | - Xingli Li
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Furong Laboratory, Xiangya School of Public Health, Central South University, 172# Tongzipo Road, Changsha, Hunan 410013, China
| | - Wen Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410078, China
| | - Fei Yang
- School of Public Health, University of South China, Hengyang, Hunan 421001, China; Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410013, China
| | - Xiangling Feng
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Furong Laboratory, Xiangya School of Public Health, Central South University, 172# Tongzipo Road, Changsha, Hunan 410013, China.
| |
Collapse
|
22
|
Cao J, Zhang H, Wang C, He L, Li Y, Wang Z, Li X, Aziz F, Yang M, Zhu X. Demethylzeylasteral inhibits osteosarcoma cell proliferation by regulating METTL17-mediated mitochondrial oxidative phosphorylation. Toxicol Appl Pharmacol 2025; 499:117348. [PMID: 40250487 DOI: 10.1016/j.taap.2025.117348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Osteosarcoma (OS) represents the most common primary bone malignancy, characterized by substantial disability and mortality, thereby underscoring the critical need for more effective therapeutic interventions to improve clinical outcomes. Demethylzeylasteral (DEM) is a bio-active compound has been reported for its anti-tumor properties through various mechanisms. Nonetheless, the specific effects of DEM on OS have yet to be fully elucidated. This study demonstrated that DEM significantly inhibited OS cell proliferation both in vitro and in vivo. Mechanistically, DEM impairs mitochondrial OXPHOS by targeting METTL17, a known regulator of mitochondrial translation, resulting in reduced ATP production. Subsequent investigations revealed that METTL17 knockdown exerts potent anti-tumor effects in OS, significantly suppressing both in vitro cell proliferation and in vivo xenograft tumor growth. Furthermore, METL17 overexpression significantly alleviated the inhibitory effects of DEM on cell proliferation, while restoring ATP production and oxygen consumption rates. These findings suggest that DEM impedes OS growth by inducing mitochondrial dysfunction through targeting METTL17, thereby highlighting a novel therapeutic strategy and potential molecular target for OS treatment.
Collapse
Affiliation(s)
- Jingyu Cao
- Institute of Pediatric Medicine, Henan Province Children's Disease Clinical Medical Research Centre, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Haotian Zhang
- Guangdong Provincial Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chengbo Wang
- Institute of Pediatric Medicine, Henan Province Children's Disease Clinical Medical Research Centre, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Lihua He
- Institute of Pediatric Medicine, Henan Province Children's Disease Clinical Medical Research Centre, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Ya Li
- Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury; Henan Provincial Outstanding Overseas Scientists Chronic Liver Injury Workshop, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zimeng Wang
- The Department of Pharmacology and Cancer, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xianxiao Li
- Accident and Emergency Care Department, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Faisal Aziz
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Minglei Yang
- The Department of Orthopedic Oncology, The Second Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Xiangzhan Zhu
- Institute of Pediatric Medicine, Henan Province Children's Disease Clinical Medical Research Centre, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China; School of Life Sciences, Zhengzhou University, Zhengzhou, China; School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
23
|
Moura JP, Oliveira PJ, Urbano AM. Mitochondria: An overview of their origin, genome, architecture, and dynamics. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167803. [PMID: 40118291 DOI: 10.1016/j.bbadis.2025.167803] [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: 12/31/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Mitochondria are traditionally viewed as the powerhouses of eukaryotic cells, i.e., the main providers of the metabolic energy required to maintain their viability and function. However, the role of these ubiquitous intracellular organelles far extends energy generation, encompassing a large suite of functions, which they can adjust to changing physiological conditions. These functions rely on a sophisticated membrane system and complex molecular machineries, most of which imported from the cytosol through intricate transport systems. In turn, mitochondrial plasticity is rooted on mitochondrial biogenesis, mitophagy, fusion, fission, and movement. Dealing with all these aspects and terminology can be daunting for newcomers to the field of mitochondria, even for those with a background in biological sciences. The aim of the present educational article, which is part of a special issue entitled "Mitochondria in aging, cancer and cell death", is to present these organelles in a simple and concise way. Complex molecular mechanisms are deliberately omitted, as their inclusion would defeat the stated purpose of the article. Also, considering the wide scope of the article, coverage of each topic is necessarily limited, with the reader directed to excellent reviews, in which the different topics are discussed in greater depth than is possible here. In addition, the multiple cell type-specific genotypic and phenotypic differences between mitochondria are largely ignored, focusing instead on the characteristics shared by most of them, with an emphasis on mitochondria from higher eukaryotes. Also ignored are highly degenerate mitochondrion-related organelles, found in various anaerobic microbial eukaryotes lacking canonical mitochondria.
Collapse
Affiliation(s)
- João P Moura
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| | - Paulo J Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
| | - Ana M Urbano
- Molecular Physical-Chemistry R&D Unit, Centre for Investigation in Environment, Genetics and Oncobiology (CIMAGO), Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
24
|
Bebawy G, Collier P, Williams PM, Burley JC, Needham D. LDLR-targeted orlistat therapeutic nanoparticles: Peptide selection, assembly, characterization, and cell-uptake in breast cancer cell lines. Int J Pharm 2025; 676:125574. [PMID: 40239877 DOI: 10.1016/j.ijpharm.2025.125574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/01/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025]
Abstract
MOTIVATION Many cancers overexpress low-density lipoprotein receptors (LDLR), facilitating cholesterol metabolism for tumour growth. Targeting LDLR offers a promising strategy for selective drug delivery. Orlistat, a fatty acid synthase (FAS) inhibitor, has shown anti-cancer potential, particularly in tumours with high FAS expression. This study introduces an LDLR-Orlistat Targeted Nanoparticles (LDLR-OTNs) to enhance cancer cell uptake via LDLR-mediated endocytosis. The objectives include synthesizing lipid-based orlistat nanoparticles, functionalizing them with an 11-mer LDLR-binding peptide, assessing uptake and cytotoxicity in three LDLR- and FAS-expressing breast cancer cell lines (BT-474, MDA MB 453, MCF-7), and comparing uptake kinetics with non-targeted nanoparticles. METHODS Orlistat nanoparticles (ONs) were synthesised via rapid solvent exchange, producing uncoated ONs, POPC-coated ONs (POPC-ONs), and LDLR-targeted ONs (LDLR-OTNs). Targeting was achieved by conjugating an 11-mer binding peptide (RLTRKRGLKLA) to DSPE-PEG5000 maleimide via click chemistry, confirmed by Ellman's test. Nanoparticles were characterised using DLS and TEM. Cellular uptake over 24 hours was assessed using fluorescence-labelled POPC-ONs and LDLR-OTNs, and uptake kinetics were analysed. Suramin-blocking studies were used to confirm LDLR-mediated uptake. A 48-hour cytotoxicity assay quantified IC50 values in the aforementioned cell lines. RESULTS TEM data showed that LDLR-OTNs (33 nm) were smaller than untargeted POPC-ONs (58 nm) and uncoated ONs (67 nm). Ellman's test confirmed > 99.2% peptide conjugation. Cellular uptake of LDLR-OTNs was rapid, with significant fluorescence by 1 hour and a kinetic plateau at 24-48 hours, with data fitting to a modified exponential model, while that of untargeted POPC-ONs had lower initial uptake, following a logistic model. Suramin blocking reduced LDLR-OTN uptake, confirming receptor-mediated entry. Cytotoxicity assays yielded IC50 values of 23.8 µM (BT-474), 25.8 µM (MDA MB 453), and 8.2 µM (MCF-7), with maximal inhibition at 48 h. CONCLUSIONS LDLR-OTNs demonstrated receptor-mediated uptake and potent cytotoxicity in LDLR- and FAS- overexpressing breast cancer cells. These findings support LDLR-targeted nanoparticles as a promising approach for delivering FAS inhibitors to LDLR-rich tumours, meriting further investigation in targeted cancer therapy development.
Collapse
Affiliation(s)
- George Bebawy
- School of Pharmacy, University of Nottingham, Nottingham, UK; Pharmaceutics Department, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Pamela Collier
- School of Pharmacy, University of Nottingham, Nottingham, UK.
| | | | | | - David Needham
- School of Pharmacy, University of Nottingham, Nottingham, UK; Department of Mechanical Engineering and Material Science, Duke University, NC, USA.
| |
Collapse
|
25
|
Kaimuangpak K, Lehtonen M, Rautio J, Weerapreeyakul N. Unraveled cancer cell survival-associated amino acid metabolism of HepG2 cells altered by Thai rat-tailed radish microgreen extract examined by untargeted LC-MS/MS analysis. Food Chem 2025; 474:143206. [PMID: 39954416 DOI: 10.1016/j.foodchem.2025.143206] [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: 07/25/2024] [Revised: 02/02/2025] [Accepted: 02/02/2025] [Indexed: 02/17/2025]
Abstract
Thai rat-tailed radish (RS) microgreens are enriched in macro- and micronutrients and phytochemicals with anticancer potential. This study investigates the antiproliferative effects of RS in the liver HepG2 cell model and untargeted liquid chromatography-mass spectrometry (LC-MS) metabolomics analysis. RS was partitioned in water and dichloromethane (DCM). DCM was collected and evaporated to yield crude extract. The extract exhibited antiproliferation with inhibitory concentrations (IC50) of 612.5 ± 24.7 μg/ml at 24 h and 568.6 ± 11.0 μg/ml at 48 h. Metabolic pathways relevant to the anticancer effects are amino acid metabolism, including (1) alanine, aspartate, and glutamate metabolism; (2) nicotinate and nicotinamide metabolism; and (3) cysteine and methionine metabolism. Significantly, glutamine was upregulated, and aspartic acid, NAD, 5'-methylthioadenosine, cystathionine, and S-adenosylhomocysteine were downregulated. This finding suggested plausible effects of RS on liver cancer cell survival and invasion activities.
Collapse
Affiliation(s)
- Karnchanok Kaimuangpak
- Graduate School (in the program of Research and Development in Pharmaceuticals), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Marko Lehtonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, 70211, Finland.
| | - Jarkko Rautio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, 70211, Finland.
| | - Natthida Weerapreeyakul
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
26
|
Kaida A, Igarashi Y, Nojima H, Nakayama M, Okada R, Takahashi R, Kobayashi H, Miura M. Uncovering cell cycle-dependent effects on cell survival in near-infrared photoimmunotherapy. Exp Cell Res 2025; 448:114570. [PMID: 40273966 DOI: 10.1016/j.yexcr.2025.114570] [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: 12/04/2024] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is an innovative cancer treatment that selectively induces cell death in cancer cells. Cetuximab-IRdye700DX (Cmab-IR700) conjugate is commonly used in NIR-PIT for head and neck squamous cell carcinoma (HNSCC) because of the frequent overexpression of epidermal growth factor receptor (EGFR) in HNSCC cells. This study examined the influence of cell cycle phases on the response and sensitivity to NIR-PIT in cell lines expressing a fluorescent ubiquitination-based cell cycle indicator (Fucci). The timing of cell death was quantified using time-lapse imaging and a clonogenic assay was used to assess cell survival. The results indicated that the timing of cell death varied among cell lines, with G1-phase cells in HSC3 and CAL33 lines showing slower cell death than those in the S/G2/M phases, whereas HeLa cells exhibited no cell cycle phase-dependent correlation. Cell rupture was predominant in HSC3 and CAL33 cells, whereas HeLa cells exhibited a combination of cell rupture and swelling. Clonogenic survival differed among the cell lines, mirroring variations in the timing of cell death. Among CAL33 and HeLa cells, G1-phase cells demonstrated greater resistance to NIR-PIT. EGFR expression levels, which varied according to cell line and cell cycle phase, were associated with sensitivity to NIR-PIT. Additionally, L-ascorbic acid-treated HeLa cells exhibited increased time to cell death and reduced NIR-PIT sensitivity, which may be due to reactive oxygen species. These findings provide information for the development of NIR-PIT strategies based on cell cycle kinetics to enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Atsushi Kaida
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan.
| | - Yuriko Igarashi
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Hitomi Nojima
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Mio Nakayama
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan; Department of Oral and Maxillofacial Surgical Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Ryuhei Okada
- Department of Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Ryosuke Takahashi
- Department of Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1088, USA
| | - Masahiko Miura
- Department of Dental Radiology and Radiation Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Japan.
| |
Collapse
|
27
|
Jiao Z, Zhang M, Ning J, Yao H, Yan X, Wu Z, Wu D, Liu Y, Zhang M, Wang L, Wang D. The oncoprotein SET promotes serine-derived one-carbon metabolism by regulating SHMT2 enzymatic activity. Proc Natl Acad Sci U S A 2025; 122:e2412854122. [PMID: 40339130 DOI: 10.1073/pnas.2412854122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/27/2025] [Indexed: 05/10/2025] Open
Abstract
Cancer cells frequently reprogram one-carbon metabolic pathways to fulfill their vigorous demands of biosynthesis and antioxidant defense for survival and proliferation. Dysfunction of oncogenes or tumor suppressor genes is critically involved in this process, but the precise mechanisms by which cancer cells actively trigger one-carbon metabolic alterations remain incompletely elucidated. Here, by using untargeted metabolomic analysis, we identify the oncoprotein SE translocation (SET) as a key regulator of one-carbon metabolism in cancer cells. SET physically interacts with mitochondrial SHMT2 and facilitates SHMT2 enzymatic activity. Loss of SET profoundly suppresses serine-derived one-carbon metabolic flux, whereas reexpression of ectopic SET leads to the opposite effect. Notably, although the presence of SHMT2 is critical for SET-mediated one-carbon metabolic alterations, the depletion of SHMT2 alone is insufficient to antagonize SET-induced tumor growth, probably due to functional compensation by its cytosolic isozyme SHMT1 upon SHMT2 knockdown. Instead, pharmacological targeting of cellular SHMT (including both SHMT1 and SHMT2) activity results in dramatic suppression of SET-induced tumor growth. Moreover, by using a Kras/Lkb1 mutation-driven lung tumor mouse model, we demonstrate that the loss of SET compromises both tumor formation and intratumoral SHMT2 enzymatic activity. Clinically, the overexpression of SET and SHMT2 is observed in lung tumors, both of which correlate with poor prognosis. Our study reveals a SET-SHMT2 axis in regulating serine-derived one-carbon metabolism and uncovers one-carbon metabolic reprogramming as a mechanism for SET-driven tumorigenesis.
Collapse
Affiliation(s)
- Zishan Jiao
- State Key Laboratory of Common Mechanism Research for Major Diseases and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Mi Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medicine, China Medical University, Shenyang 110122, China
| | - Jingyuan Ning
- State Key Laboratory of Common Mechanism Research for Major Diseases and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Han Yao
- State Key Laboratory of Common Mechanism Research for Major Diseases and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Xiaojun Yan
- State Key Laboratory of Common Mechanism Research for Major Diseases and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Zhen Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Dexuan Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yajing Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Meng Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Lin Wang
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Donglai Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
28
|
Zhu S, Sun C, Cai Z, Wu J, Han X, Wang J, Wang C. Multifunctional nanoparticle-mediated targeting of metabolic reprogramming and DNA damage response pathways to treat drug-resistant triple-negative breast cancer. J Control Release 2025; 381:113601. [PMID: 40054629 DOI: 10.1016/j.jconrel.2025.113601] [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: 12/07/2024] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Multi-drug resistance and immunosuppressive triple-negative breast cancer (TNBC) is triggered by the Warburg effect, which promotes homologous recombination repair (HRR) and upregulates expression of P-glycoprotein (P-gp), in turn preventing DNA damage from chemotherapy and creating an immunosuppressive microenvironment. It is therefore of clinical relevance to develop an effective delivery system that targets metabolic reprogramming and DNA damage response pathways for the treatment of drug-resistant TNBC. Herein, a P-gp-inhibiting and GSH-responsive multifunctional drug carrier targeting integrin αvβ3 was synthesised for the delivery of Lonidamine-prodrug (M1, glycolysis inhibitor) and Senaparib (Se, Poly [ADP-ribose] polymerase inhibitor). The nanodrug delivery system (iPR@M1/Se nanoparticles) exhibit effective tumour penetration and P-gp inhibition, effectively inducing DNA damage and apoptosis in Olaparib-resistant TNBC cells in vitro, as well as a higher tumour inhibitory rate compared with that of Se (81.82 % ± 2.31 % vs 43.91 % ± 4.65 %) in vivo. Mechanistically, iPR@M1/Se nanoparticles not only reshaped the immunosuppressive microenvironment resulting from tumour glycolysis, but also downregulated the expression of HRR-related protein, fostering the cytoplasmic accumulation of DNA damage fragments, which induced activation of the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon gene (STING) pathway. Experimental results show that iPR@M1/Se nanoparticles effectively promote dendritic cell maturation and T lymphocyte activation, which elicits long-term immune memory responses, and prevents tumour recurrence and lung metastasis. Therefore, these multifunctional nanoparticles have great potential and provide a clinically relevant and valuable option for Olaparib-resistant TNBC.
Collapse
Affiliation(s)
- Sifeng Zhu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Chao Sun
- Institute of Medical Science, Central Research Laboratory, the Second Hospital of Shandong University, 250033, China
| | - Zimin Cai
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jibin Wu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xu Han
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jue Wang
- Institute of Medical Sciences, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China.
| | - Cheng Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
29
|
Mihalas AB, Arora S, O'Connor SA, Feldman HM, Cucinotta CE, Mitchell K, Bassett J, Kim D, Jin K, Hoellerbauer P, Delegard J, Ling M, Jenkins W, Kufeld M, Corrin P, Carter L, Tsukiyama T, Aronow B, Plaisier CL, Patel AP, Paddison PJ. KAT5 regulates neurodevelopmental states associated with G0-like populations in glioblastoma. Nat Commun 2025; 16:4327. [PMID: 40346033 DOI: 10.1038/s41467-025-59503-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Quiescence cancer stem-like cells may play key roles in promoting tumor cell heterogeneity and recurrence for many tumors, including glioblastoma (GBM). Here we show that the protein acetyltransferase KAT5 is a key regulator of transcriptional, epigenetic, and proliferative heterogeneity impacting transitions into G0-like states in GBM. KAT5 activity suppresses the emergence of quiescent subpopulations with neurodevelopmental progenitor characteristics, while promoting GBM stem-like cell (GSC) self-renewal through coordinately regulating E2F- and MYC- transcriptional networks with protein translation. KAT5 inactivation significantly decreases tumor progression and invasive behavior while increasing survival after standard of care. Further, increasing MYC expression in human neural stem cells stimulates KAT5 activity and protein translation, as well as confers sensitivity to homoharringtonine, to similar levels to those found in GSCs and high-grade gliomas. These results suggest that the dynamic behavior of KAT5 plays key roles in G0 ingress/egress, adoption of quasi-neurodevelopmental states, and aggressive tumor growth in gliomas.
Collapse
Affiliation(s)
- Anca B Mihalas
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Sonali Arora
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Samantha A O'Connor
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Heather M Feldman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Christine E Cucinotta
- College of Arts and Sciences, Department of Molecular Genetics, Ohio State University, Columbus, OH, 43210, USA
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Kelly Mitchell
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - John Bassett
- Department of Medicine, Karolinska Institute, Huddinge, Sweden
| | - Dayoung Kim
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Kang Jin
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Pia Hoellerbauer
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Jennifer Delegard
- Department of Neurosurgery, University of Washington, Seattle, WA, 98195, USA
| | - Melissa Ling
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195, USA
| | - Wesley Jenkins
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195, USA
| | - Megan Kufeld
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Philip Corrin
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Lucas Carter
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Toshio Tsukiyama
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Bruce Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Christopher L Plaisier
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Anoop P Patel
- Department of Neurosurgery, Duke University, Durham, NC, 27710, USA.
- Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC, 27710, USA.
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, 27710, USA.
| | - Patrick J Paddison
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
30
|
Kristjansen KA, Djebbara-Bozo N, Nanthan KR, Bønnelykke-Behrndtz ML. Repurposing tranexamic acid as an anticancer drug: a systematic review and meta-analysis. J Cancer Res Clin Oncol 2025; 151:157. [PMID: 40343490 DOI: 10.1007/s00432-025-06185-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 05/11/2025]
Abstract
PURPOSE Drug repurposing may be an efficient strategy for identifying new cancer treatments. Tranexamic acid (TXA), an antifibrinolytic agent that affects the plasminogen-plasmin pathway, may have potential anticancer effects by influencing tumor cell proliferation, angiogenesis, inflammation, immune response, and tissue remodeling-all crucial processes contributing to tumor progression and metastasis. OBJECTIVE Evaluate TXA's anticancer effects across in vitro, animal, and clinical studies to assess its potential as a repurposed cancer drug. METHODS The study was designed as a PRISMA-compliant systematic review and meta-analysis. The literature search was conducted in MEDLINE, EMBASE, Web of Science, and the Cochrane Library. In vitro, animal, and clinical studies investigating the anticancer effects of TXA or epsilon-aminocaproic acid (EACA) were included. Animal and clinical studies were critically appraised, and studies with a low risk of bias were included in the meta-analysis. RESULTS Of 4367 identified records, 38 articles were included, collectively reporting findings from 41 in vitro studies, 34 animal studies (n = 843 animals), and seven clinical studies (n = 91 patients). The meta-analysis included nine animal studies and showed a tumor growth reduction in animals treated with TXA compared to controls with a standardized mean difference of - 1.0 (95%CI - 1.5; - 0.4) (p = 0.0002). Equivalently, the majority of in vitro studies reported reduced proliferation, viability, and invasiveness in TXA-exposed tumor cell lines. The clinical studies were considerably susceptible to bias, rendering any conclusions futile. CONCLUSIONS TXA shows promise as a repurposed cancer drug, revealing an overall reduction in tumor growth, viability, and invasiveness in animal and in vitro studies.
Collapse
Affiliation(s)
- Karoline Assifuah Kristjansen
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
- Department of Plastic and Breast Surgery, Aalborg University Hospital, Søndre Skovvej 3, 9000, Aalborg, Denmark.
| | - Nulvin Djebbara-Bozo
- Department of Plastic Surgery and Burns Treatment, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Kumanan Rune Nanthan
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Aalborg University Hospital, Søndre Skovvej 3, 9000, Aalborg, Denmark
| | - Marie Louise Bønnelykke-Behrndtz
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
- Department of Plastic and Breast Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| |
Collapse
|
31
|
Jaiswal N, Mahata N, Chanda N. Nanogold-albumin conjugates: transformative approaches for next-generation cancer therapy and diagnostics. NANOSCALE 2025; 17:11191-11220. [PMID: 40237258 DOI: 10.1039/d4nr05279j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Nanogold-albumin conjugates have garnered significant attention as a highly adaptable theranostic platform, capable of delivering a wide range of therapeutics, from small-molecule drugs to larger biomolecules, while offering promising applications for monitoring and managing cancer. The remarkable theranostic capabilities of these conjugates stem from the combined strengths of gold and albumin, which provide low toxicity, a large surface area, customizable surface chemistry, and unique optical properties, all contributing to their potential in cancer therapy. This review delves into the design and development of two primary types of nanogold-albumin conjugate: supramolecular albumin-coated gold nanoparticles (GNP-BSA/HSA) and albumin-templated ultra-small gold nanoclusters (GNC-BSA/HSA). Each strategy offers distinct advantages, enabling the fine-tuning of conjugate properties to optimize therapeutic delivery and facilitate cancer-specific bio-sensing. The integration of gold and albumin further improves biocompatibility, extends circulation time, and enhances tumor targeting, making these conjugates an attractive option for cancer treatment. The review also focuses on the refinement of surface chemistry to achieve precise targeting of cancer cells, as well as the challenges and future prospects for advancing nanogold-albumin systems in clinical applications.
Collapse
Affiliation(s)
- Namita Jaiswal
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nripen Chanda
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
| |
Collapse
|
32
|
Chen Y, Huang X, Hu R, Lu E, Luo K, Yan X, Zhang Z, Ma Y, Zhang M, Sha X. Inhalable biomimetic polyunsaturated fatty acid-based nanoreactors for peroxynitrite-augmented ferroptosis potentiate radiotherapy in lung cancer. J Nanobiotechnology 2025; 23:338. [PMID: 40340938 PMCID: PMC12060495 DOI: 10.1186/s12951-025-03409-8] [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: 01/26/2025] [Accepted: 04/19/2025] [Indexed: 05/10/2025] Open
Abstract
The limited efficacy and poor tumor accumulation remain crucial challenges for radiotherapy against lung cancer. To address these limitations, we rationally developed a polyunsaturated fatty acid (PUFA)-based nanoreactor (DHA-N@M) camouflaged with macrophage cell membrane to improve tumoral distribution and achieve peroxynitrite-augment ferroptosis for enhanced radiotherapy against lung cancer. After nebulization, the nanoreactors exhibited superior pulmonary accumulation in orthotopic lung cancer-bearing mice, with 70-fold higher than intravenously injected nanoreactors at 12 h post-administration, and distributed deeply in the tumors. DHA-N@M selectively released nitric oxide (NO) in glutathione (GSH)-enriched tumor cells, with consumption of GSH and subsequent inactivation of glutathione peroxidase 4 (GPX4). Under radiation, NO reacted with radiotherapy-induced reactive oxygen species (ROS) to generate peroxynitrite (ONOO-), resulting in redox homeostasis disruption. Combined with docosahexaenoic acid (DHA)-induced lipid metabolism disruption, overwhelming ferroptosis was induced both in vitro and in vivo. Notably, DHA-N@M mediated ferroptosis-radiotherapy significantly suppressed tumor growth with a 93.91% inhibition in orthotopic lung cancer models. Therefore, this design provides a nebulized ferroptosis-radiotherapy strategy for lung cancer.
Collapse
Affiliation(s)
- Yiting Chen
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Xueli Huang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Ruining Hu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Enhao Lu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Kuankuan Luo
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Xin Yan
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Zhiwen Zhang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Yan Ma
- Department of Pharmacy, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
| | - Minghe Zhang
- Naval Medical Center, Naval Medical University, Shanghai, Shanghai, 200052, China.
| | - Xianyi Sha
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China.
- Quzhou Fudan Institute, 108 Minjiang Avenue, Kecheng District, Quzhou, Zhejiang Province, 324002, China.
| |
Collapse
|
33
|
Wu C, Luo D, Shi B, Chen S, Sun C, He Z, Yu C. Sciellin inhibits senescence and promotes pancreatic cancer progress by activating the notch signaling pathway. Sci Rep 2025; 15:16133. [PMID: 40341648 PMCID: PMC12062315 DOI: 10.1038/s41598-025-88265-0] [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: 06/21/2024] [Accepted: 01/28/2025] [Indexed: 05/10/2025] Open
Abstract
Pancreatic cancer (PC) incidence is increasing annually globally, and the five-year survival rate of patients with PC is approximately 10%. Cellular senescence is a regulatory mechanism against cancer that prevents tumor development by inhibiting the proliferation of damaged or abnormal cells. However, the mechanisms underlying cellular senescence in PC is unclear. Sciellin (SCEL) is a precursor protein of the cornified envelope predominantly enriched in epithelial cells. Previous studies have discovered potential links between SCEL and cellular senescence through bioinformatics analysis. Therefore, the specific role of SCEL in cellular senescence and the malignant features of PC are unclear. In vivo and in vitro assays were performed to investigate the role of SCEL in PC cell senescence, proliferation, invasion, and metastasis. Gene set enrichment analysis was used to identify the Notch signaling pathways activated by SCEL, and coimmunoprecipitation was used to detect proteins that interact with SCEL. The results revealed that SCEL was significantly upregulated in PC tissues and cell models and was correlated with poor clinical outcomes. Further investigation revealed that the interaction between SCEL and Jagged-1 promotes the activation of the Notch signaling pathway, effectively inhibiting the senescence of PC cells while enhancing their proliferation, invasion, and metastatic capabilities. Therefore, SCEL is a potential therapeutic target for PC.
Collapse
Affiliation(s)
- Changhao Wu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Dan Luo
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
- Department of Hepatobiliary Surgery, People's Hospital of the Guizhou Province, Guiyang, 550003, China
| | - Binbin Shi
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Shiyu Chen
- Department of Hepatic-Biliary-Pancreatic Surgery, Medical School, South China Hospital, Shenzhen University, Shenzhen, 518116, China
| | - Chengyi Sun
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China.
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China.
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China.
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China.
| | - Chao Yu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China.
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China.
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China.
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China.
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
34
|
Räisänen M, Kaasinen E, Jäntti M, Taira A, Siili E, Bützow R, Heikinheimo O, Pasanen A, Karhu A, Berta DG, Välimäki N, Aaltonen LA. Chromatin state origins of uterine leiomyoma. Nat Commun 2025; 16:4307. [PMID: 40341524 PMCID: PMC12062214 DOI: 10.1038/s41467-025-59646-w] [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: 09/25/2024] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Aberrations in the regulatory genome play a pivotal role in population-level disease predisposition. Annotation of the regulatory regions using appropriate primary tissues - instead of cell lines affected by selection and other confounding factors - could shed new light into mechanisms underlying common conditions. We test this approach in uterine leiomyomas, highly prevalent benign neoplasms of the myometrium, by creating 15-state chromatin annotations for myometrium and uterine leiomyomas. Integration with RNA-seq, ATAC-seq, HiChIP and methylation data enables us to compare the epigenomes of myometrium and ULs with distinct driver mutations, highlighting the role of bivalent regions in the neoplastic process. Subsequently, a genome wide association study meta-analysis is performed, using three different cohorts. Disease association loci are enriched at active chromatin, especially at enhancers, and harbor tumor- and driver mutation-specific chromatin states. At SATB2 locus we show the effect of the risk genotype already in the normal tissue. Integration of genome-wide association studies and deep regulatory genomics data from the correct tissue type represents a powerful approach in understanding population-level disease predisposition.
Collapse
Affiliation(s)
- Maritta Räisänen
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Eevi Kaasinen
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Maija Jäntti
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Aurora Taira
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Emma Siili
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ralf Bützow
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Oskari Heikinheimo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Annukka Pasanen
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Auli Karhu
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Davide G Berta
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Niko Välimäki
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Lauri A Aaltonen
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
35
|
Tian Y, Ma L, Liu S, Bai X, Shah N, Zhang L, Wang X, Zhang Y, Ding X. The prognostic value of AST-lymphocyte ratio index in liver cancer patients treated with TACE: a systematic review and single-center retrospective study. BMC Gastroenterol 2025; 25:348. [PMID: 40340835 PMCID: PMC12063415 DOI: 10.1186/s12876-025-03949-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND AND AIMS AST-lymphocyte ratio index (ALRI) has been proposed as a potentially prognostic indicator of liver cancer patients underwent transcatheter arterial chemoembolization (TACE) in studies, but the numbers were small and the results were controversial. In this study, we systematically assessed the prognostic value of ALRI in liver cancer patients treated with TACE by integrating meta-analysis with single-center clinical analysis. METHODS We conducted a systematic literature search across multiple databases and evaluated the quality of included studies using the Newcastle-Ottawa Scale. We employed a fixed-effect model to calculate the pooled hazard ratio (HR) and 95% confidence interval (CI). Publication bias were evaluated using funnel plot, Begg's and Egger's tests. Concurrently, we integrated clinical data from 127 HCC patients treated with TACE at our center, employed X-tile software to ascertain the optimal cutoff value for ALRI, and analyzed the relationship between ALRI and clinical characteristics as well as overall survival (OS), using chi-square tests, Kaplan-Meier survival curves, and Cox proportional hazards models. RESULTS The meta-analysis included 7 studies, and the pooled hazard ratio (HR) indicated that elevated ALRI was significantly associated with poorer OS in liver cancer patients underwent TACE (HR = 1.75, 95% CI: 1.46-2.1, P<0.01), with no significant heterogeneity (P = 0.542, I2 = 0.00%). Clinical analysis of 127 patients further supported this finding, with patients in the high ALRI group showed significantly lower OS compared to those in the low ALRI group (1-year OS rate: 96.7% vs. 87.9%, 2-year OS rate: 61.5% vs. 42.7%; C2 = 28.006, P<0.01). Multivariate Cox regression analysis revealed that number of tumors, tumor size and ALRI were all independent prognostic factors for OS (ALRI HR = 6.456, 95%CI: 2.247-18.55, P < 0.01). CONCLUSIONS An increase in ALRI may serve as an independent prognostic indicator of poor outcomes in liver cancer patients undergoing TACE. While it offers benefits such as being non-invasive and cost-effective, further large-scale, multicenter, prospective studies are essential to validate the efficacy of ALRI and establish standardized cutoff values for clinical application.
Collapse
Affiliation(s)
- Yali Tian
- Department of Infectious Disease, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, No. 301, Zhengyuan North Street, Jinfeng District, Yinchuan, Ningxia Hui Autonomous Region, 750021, China
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Lina Ma
- Department of Infectious Disease, General Hospital of Ningxia Medical University, No. 804, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Shuaiwei Liu
- Department of Infectious Disease, General Hospital of Ningxia Medical University, No. 804, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Xiaoyang Bai
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Nawaz Shah
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Le Zhang
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Xia Wang
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Yuxi Zhang
- Department of Infectious Disease, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, No. 301, Zhengyuan North Street, Jinfeng District, Yinchuan, Ningxia Hui Autonomous Region, 750021, China.
| | - Xiangchun Ding
- Department of Infectious Disease, General Hospital of Ningxia Medical University, No. 804, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China.
- Infectious Disease Clinical Research Center of Ningxia, 804 Shengli Street, Xingqing District, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
36
|
Techachakrit J, Malik AA, Pisitkun T, Sriswasdi S. Potential shared neoantigens from pan-cancer transcript isoforms. Sci Rep 2025; 15:15886. [PMID: 40335513 PMCID: PMC12059137 DOI: 10.1038/s41598-025-00817-6] [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: 10/15/2024] [Accepted: 04/30/2025] [Indexed: 05/09/2025] Open
Abstract
Isoform switching in cancer is a prevalent phenomenon with significant implications for immunotherapy, as actionable neoantigens derived from these cancer-specific events would be applicable to broad categories of patients, reducing the necessity for personalized treatments. By integrating five large-scale transcriptomic datasets comprising over 19,500 samples across 29 cancer and 54 normal tissue types, we identified cancer-associated isoform switching events common to multiple cancer types, several of which involve genes with established mechanistic roles in oncogenesis. The presence of neoantigen-containing peptides derived from these transcripts was confirmed in broad cancer and normal tissue proteome datasets and the binding affinity of predicted neoantigens to the human leukocyte antigen (HLA) complex via molecular dynamics simulations. The study presents strong evidence that isoform switching in cancer is a significant source of actionable neoantigens that have the capability to trigger an immune response. These findings suggest that isoform switching events could potentially be leveraged for broad immunotherapeutic strategies across various cancer types.
Collapse
Affiliation(s)
- Jirapat Techachakrit
- Center of Excellence in Computational Molecular Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Aijaz Ahmad Malik
- Center of Excellence in Computational Molecular Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center for Artificial Intelligence in Medicine, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sira Sriswasdi
- Center of Excellence in Computational Molecular Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center for Artificial Intelligence in Medicine, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
37
|
Wang Z, Guo W, Zhang X, Wei Y, Zhang W, Du N, Li C, Wu X, Yi F, Zhou T, Dong X, Guo Q, Xu H, Wang E, Li N, Cheng R, Li Z, Song X, Sun Y, Sun X, Cao L. Tumor microenvironment-associated oxidative stress impairs SIRT1 secretion to suppress anti-tumor immune response. Cell Rep 2025; 44:115679. [PMID: 40343797 DOI: 10.1016/j.celrep.2025.115679] [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: 05/15/2024] [Revised: 11/10/2024] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
Sirtuin-1 (SIRT1) is a classical histone deacetylase well known for its roles in intracellular pathways such as energy metabolism, DNA damage response, and genome stability maintenance. We report that SIRT1 can be secreted into the tumor microenvironment (TME) through an unconventional protein secretion pathway, effectively inhibiting tumor growth. However, under the stressful conditions of the TME, SIRT1 undergoes increased methylation, which impedes its secretion. Consequently, tumor-infiltrating M2 macrophages are unable to acquire sufficient SIRT1 from the TME, resulting in a significant decrease in SIRT1 levels within these cells. This SIRT1 decline leads to elevated expression of programmed cell death ligand 1 (PD-L1) on M2 macrophages, which in turn contributes to CD8+ T cell exhaustion through the programmed cell death protein 1/PD-L1 interaction pathway. These findings unveil the multifaceted roles and regulatory mechanisms of SIRT1 within the complex TME, providing deeper insights that significantly enhance our understanding of tumor immune-evasion strategies.
Collapse
Affiliation(s)
- Zhuo Wang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Wendong Guo
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaowen Zhang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Yufei Wei
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning 110122, China
| | - Wanying Zhang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Ning Du
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Chunlu Li
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xuan Wu
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Fei Yi
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Tingting Zhou
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiang Dong
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning 110122, China
| | - Qiqiang Guo
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Hongde Xu
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Erli Wang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Na Li
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Rong Cheng
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Ziwei Li
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaoyu Song
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China.
| | - Yingxian Sun
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning 110122, China.
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning 110122, China.
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
38
|
Zhang M, Liu C, Tu J, Tang M, Ashrafizadeh M, Nabavi N, Sethi G, Zhao P, Liu S. Advances in cancer immunotherapy: historical perspectives, current developments, and future directions. Mol Cancer 2025; 24:136. [PMID: 40336045 PMCID: PMC12057291 DOI: 10.1186/s12943-025-02305-x] [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: 01/05/2025] [Accepted: 03/15/2025] [Indexed: 05/09/2025] Open
Abstract
Cancer immunotherapy, encompassing both experimental and standard-of-care therapies, has emerged as a promising approach to harnessing the immune system for tumor suppression. Experimental strategies, including novel immunotherapies and preclinical models, are actively being explored, while established treatments, such as immune checkpoint inhibitors (ICIs), are widely implemented in clinical settings. This comprehensive review examines the historical evolution, underlying mechanisms, and diverse strategies of cancer immunotherapy, highlighting both its clinical applications and ongoing preclinical advancements. The review delves into the essential components of anticancer immunity, including dendritic cell activation, T cell priming, and immune surveillance, while addressing the challenges posed by immune evasion mechanisms. Key immunotherapeutic strategies, such as cancer vaccines, oncolytic viruses, adoptive cell transfer, and ICIs, are discussed in detail. Additionally, the role of nanotechnology, cytokines, chemokines, and adjuvants in enhancing the precision and efficacy of immunotherapies were explored. Combination therapies, particularly those integrating immunotherapy with radiotherapy or chemotherapy, exhibit synergistic potential but necessitate careful management to reduce side effects. Emerging factors influencing immunotherapy outcomes, including tumor heterogeneity, gut microbiota composition, and genomic and epigenetic modifications, are also examined. Furthermore, the molecular mechanisms underlying immune evasion and therapeutic resistance are analyzed, with a focus on the contributions of noncoding RNAs and epigenetic alterations, along with innovative intervention strategies. This review emphasizes recent preclinical and clinical advancements, with particular attention to biomarker-driven approaches aimed at optimizing patient prognosis. Challenges such as immunotherapy-related toxicity, limited efficacy in solid tumors, and production constraints are highlighted as critical areas for future research. Advancements in personalized therapies and novel delivery systems are proposed as avenues to enhance treatment effectiveness and accessibility. By incorporating insights from multiple disciplines, this review aims to deepen the understanding and application of cancer immunotherapy, ultimately fostering more effective and widely accessible therapeutic solutions.
Collapse
Affiliation(s)
- Meiyin Zhang
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chaojun Liu
- Department of Breast Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Jing Tu
- Department of Pulmonary and Critical Care Medicine, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8 V 1P7, Canada
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR) Yong Loo Lin, School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Peiqing Zhao
- Translational Medicine Center, Zibo Central Hospital Affiliated to Binzhou Medical University, No. 54 Communist Youth League Road, Zibo, China.
| | - Shijian Liu
- Department of General Medicine, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, 150081, China.
| |
Collapse
|
39
|
Okonechnikov K, Joshi P, Körber V, Rademacher A, Bortolomeazzi M, Mallm JP, Vaillant J, da Silva PBG, Statz B, Sepp M, Sarropoulos I, Yamada T, Wittmann A, Schramm K, Blattner-Johnson M, Fiesel P, Jones B, Jäger N, Milde T, Pajtler KW, van Tilburg CM, Witt O, Bochennek K, Weber KJ, Nonnenmacher L, Reimann C, Ghasemi DR, Schüller U, Mynarek M, Rutkowski S, Jones DTW, Korshunov A, Rippe K, Westermann F, Thongjuea S, Höfer T, Kaessmann H, Kutscher LM, Pfister SM. Oncogene aberrations drive medulloblastoma progression, not initiation. Nature 2025:10.1038/s41586-025-08973-5. [PMID: 40335697 DOI: 10.1038/s41586-025-08973-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/03/2025] [Indexed: 05/09/2025]
Abstract
Despite recent advances in understanding disease biology, treatment of group 3/4 medulloblastoma remains a therapeutic challenge in paediatric neuro-oncology1. Bulk-omics approaches have identified considerable intertumoural heterogeneity in group 3/4 medulloblastoma, including the presence of clear single-gene oncogenic drivers in only a subset of cases, whereas in most cases, large-scale copy number aberrations prevail2,3. However, intratumoural heterogeneity, the role of oncogene aberrations, and broad copy number variation in tumour evolution and treatment resistance remain poorly understood. To dissect this interplay, we used single-cell technologies (single-nucleus RNA sequencing (snRNA-seq), single-nucleus assay for transposase-accessible chromatin with high-throughput sequencing (snATAC-seq) and spatial transcriptomics) on a cohort of group 3/4 medulloblastoma with known alterations in the oncogenes MYC, MYCN and PRDM6. We show that large-scale chromosomal aberrations are early tumour-initiating events, whereas the single-gene oncogenic events arise late and are typically subclonal, but MYC can become clonal upon disease progression to drive further tumour development and therapy resistance. Spatial transcriptomics shows that the subclones are mostly interspersed across tumour tissue, but clear segregation is also present. Using a population genetics model, we estimate medulloblastoma initiation in the cerebellar unipolar brush cell lineage starting from the first gestational trimester. Our findings demonstrate how single-cell technologies can be applied for early detection and diagnosis of this fatal disease.
Collapse
Affiliation(s)
- Konstantin Okonechnikov
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Piyush Joshi
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Körber
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Anne Rademacher
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and Bioquant, Heidelberg, Germany
| | | | - Jan-Philipp Mallm
- Single-cell Open Lab, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Vaillant
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Patricia Benites Goncalves da Silva
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Britta Statz
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Mari Sepp
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Ioannis Sarropoulos
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Tetsuya Yamada
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Andrea Wittmann
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kathrin Schramm
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mirjam Blattner-Johnson
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petra Fiesel
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- CCU Neuropathology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Barbara Jones
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Natalie Jäger
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Till Milde
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Kristian W Pajtler
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Cornelis M van Tilburg
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Konrad Bochennek
- Frankfurt University Hospital, Goethe University, Frankfurt, Germany
| | - Katharina Johanna Weber
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT) Frankfurt, Frankfurt am Main, Germany
| | | | | | - David R Ghasemi
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
- Department of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Mynarek
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Rutkowski
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrey Korshunov
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- CCU Neuropathology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and Bioquant, Heidelberg, Germany
| | - Frank Westermann
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Supat Thongjuea
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Henrik Kaessmann
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena M Kutscher
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany.
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Stefan M Pfister
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany.
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
40
|
Zhou P, Huang R, Cheng Y, Yang Y, Qian D, Ming X, Wang AZ, Chen X, Min Y. Nanotherapeutic Wee1 Inhibition Sensitizes Tumor Ferroptosis to Promote Cancer Immunotherapy and Abscopal Effect. ACS NANO 2025; 19:16307-16326. [PMID: 40263774 DOI: 10.1021/acsnano.4c13218] [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: 04/24/2025]
Abstract
The major issue with cancer immunotherapy is the low response rate. So, development of therapeutics enhancing immune responses is an urgent need. Tumor ferroptosis could produce immunogenic cancer cell death, which may improve cancer immunotherapy. However, current ferroptosis inducers may be limited to specific genetic backgrounds of cancer cells. Therefore, sensitization to ferroptosis inducers has also been highly pursued. Here, we found that Wee1 expression was negatively associated with drug sensitivity and positively correlated with an immunosuppressive microenvironment. Further investigation demonstrated that Wee1 inhibition could result in changes of ferroptosis and iron ion homeostasis, regardless of p53 status. Our in vitro results demonstrated the underlying mechanism that Wee1 inhibition primed cancer cells to ferroptosis through mitochondria reactive oxygen species and labile iron-dependent pathways. In order to decrease side effects, we developed an acidic responsive nanoformulation of the Wee1 inhibitor, which can sensitize tumor ferroptosis in vivo and also improve the response of cancer immunotherapy. Combining immunotherapy, nanotherapeutic Wee1 inhibition also produced abscopal effect with up to 55% mice cured that has not been seen before. In summary, nanotherapeutic Wee1 inhibition sensitized ferroptosis to enhance cancer immunotherapy and abscopal effect.
Collapse
Affiliation(s)
- Peijie Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Ruijie Huang
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Yong Cheng
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yidong Yang
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- School of Physical Sciences and Ion Medical Research Institute, University of Science and Technology of China, Hefei 230026, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xin Ming
- Department of Cancer Biology and Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston Salem, North Carolina 27157, United States
| | - Andrew Z Wang
- Department of Radiation Oncology, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Yuanzeng Min
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
- Department of Endocrinology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
41
|
Bai J, Xiao R, Jiang D, Luo X, Tang Y, Cui M, You L, Zhao Y. Sialic Acids: Sweet Modulators Fueling Cancer Cells and Domesticating the Tumor Microenvironment. Cancer Lett 2025:217773. [PMID: 40339953 DOI: 10.1016/j.canlet.2025.217773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/23/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Tumor microenvironment (TME) can shift towards either immune activation or immunosuppression, influenced by various factors. Recent studies have underscored the pivotal role of sialic acids, a group of monosaccharides with a 9-carbon backbone, in modulating the TME. Aberrant expression or abnormal addition of sialic acids to the surface of cancer cells and within the tumor stroma has been identified as a key contributor to tumor progression. Abnormal sialylation on cancer cell surfaces can inhibit apoptosis, enhance cell proliferation, and facilitate metastasis. Notably, recent findings suggest that dysregulated sialic acid expression in the TME actively contributes to shaping an immunosuppressive niche by reducing the population of anti-tumor immune cells and impairing immune cell function. The mechanisms by which sialic acids foster immune escape and shape the immunosuppressive TME have been partially unraveled, particularly through interactions with sialic acid receptors on immune cells. Importantly, several sialic acid-targeted therapies are currently advancing into clinical trials, offering promising prospects for clinical translation. This dysregulated sialylation represents a significant opportunity for molecular diagnostics and therapeutic interventions in oncology. Targeting aberrant sialylation or disrupting the interaction between sialic acids and their receptors offers potential strategies to reprogram the TME towards an anti-tumor phenotype, thereby facilitating the advancement of innovative cancer therapies.
Collapse
Affiliation(s)
- Jialu Bai
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Decheng Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiyuan Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuemeng Tang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ming Cui
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
42
|
Zhu S, Liu Y, Chen H, Zhu X, Liu X, Xu K, Sang Y, Shang L, Chong W, Li L. Mechanism and Therapeutic Progress of One-Carbon Metabolic Key Enzyme: Serine Hydroxymethyltransferase 2 in Cancer. Clin Med Insights Oncol 2025; 19:11795549251331755. [PMID: 40337354 PMCID: PMC12056339 DOI: 10.1177/11795549251331755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/13/2025] [Indexed: 05/09/2025] Open
Abstract
Serine hydroxymethyltransferase 2 (SHMT2) is a crucial mitochondrial enzyme in 1-carbon (1C) metabolism. It catalyzes the conversion of serine to glycine, generating 1C units essential for purine and pyrimidine synthesis, thereby supporting DNA replication and repair. Abnormally high expression is associated with malignant progression and treatment tolerance in various cancers. This review systematically summarizes the functions of SHMT2 in different types of cancer, underscoring on its roles in metabolism, immune microenvironment, and key signaling pathways (PI3K/AKT/mTOR, JAK-STAT, etc.) and outlines its epigenetic regulation and posttranslational modification mechanisms. Compared with the existing research, we focused on the latest regulatory mechanisms of SHMT2 and its potential in cancer treatment, such as the development and application of small-molecule inhibitors (SHIN2 and AGF347).
Collapse
Affiliation(s)
- Siqiang Zhu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuan Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Chen
- Clinical Research Center of Shandong University, Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Xingyu Zhu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xinyu Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Kang Xu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yaodong Sang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
43
|
Turvey GL, López de Alba E, Stewart E, Cook H, Alalti A, Gawne RT, Ainscough JFX, Mason AS, Coverley D. Epigenetic deprogramming by disruption of CIZ1-RNA nuclear assemblies in early-stage breast cancers. J Cell Biol 2025; 224:e202409123. [PMID: 40067149 PMCID: PMC11895699 DOI: 10.1083/jcb.202409123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/11/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
CIZ1 is part of the RNA-dependent supramolecular assemblies that form around the inactive X-chromosome (Xi) in female cells and smaller assemblies throughout the nucleus in both sexes. Here, we show that CIZ1 C-terminal anchor domain (AD) is elevated in human breast tumor transcriptomes, even at stage I. Elevation correlates with deprotection of chromatin and upregulation of lncRNA-containing gene clusters in ∼10 Mb regions enriched in cancer-associated genes. We modeled the effect of AD on endogenous CIZ1-Xi assemblies and observed dominant-negative interference with their reformation after mitosis, leading to abnormal assemblies similar to those in breast cancer cells, and depletion of H2AK119ub1, H3K27me3, and Xist. Consistent alterations in gene expression were evident across the genome, showing that AD-mediated interference has a destabilizing effect, likely by unscheduled exposure of underlying chromatin to modifying enzymes. The data argue for a dominant, potent, and rapid effect of CIZ1 AD that can deprogram gene expression patterns and which may predispose incipient tumors to epigenetic instability.
Collapse
Affiliation(s)
- Gabrielle L. Turvey
- Mammalian Cell Cycle Research Group, Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - Ernesto López de Alba
- Mammalian Cell Cycle Research Group, Department of Biology, University of York, York, UK
| | - Emma Stewart
- Mammalian Cell Cycle Research Group, Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - Heather Cook
- Mammalian Cell Cycle Research Group, Department of Biology, University of York, York, UK
| | - Ahmad Alalti
- Mammalian Cell Cycle Research Group, Department of Biology, University of York, York, UK
| | - Richard T. Gawne
- York Biomedical Research Institute, University of York, York, UK
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York, UK
| | - Justin F.-X. Ainscough
- Mammalian Cell Cycle Research Group, Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - Andrew S. Mason
- York Biomedical Research Institute, University of York, York, UK
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York, UK
| | - Dawn Coverley
- Mammalian Cell Cycle Research Group, Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| |
Collapse
|
44
|
Needleman RA, Thai AA. The complexities of PD-L1 expression as an indicator of immunotherapy outcomes. Immunotherapy 2025:1-4. [PMID: 40325617 DOI: 10.1080/1750743x.2025.2500908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025] Open
Affiliation(s)
| | - Alesha A Thai
- Department of Medical Oncology, Austin Hospital, Heidelberg, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
45
|
Fang X, Ruan Y, Yin X, Wang J, Chen C, Hu Y, Wang H, Pi J, Xu Y. The role of SLC7A11 in arsenite-induced oncogenic phenotypes of human bronchial epithelial cells: A metabolic perspective. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 376:126381. [PMID: 40334737 DOI: 10.1016/j.envpol.2025.126381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/14/2025] [Accepted: 05/04/2025] [Indexed: 05/09/2025]
Abstract
Chronic arsenic exposure enhances the probability of lung cancer with the underlying mechanisms remain unknown. Glutamine-driven synthetic metabolism, including nucleotide synthesis, amino acid production, TCA cycle replenishment, glutathione synthesis, and lipid biosynthesis, is crucial for both cancer initiation and progression. This study demonstrated that chronic exposure to 0.1 μM arsenite for as long as 36 weeks induced malignant transformation in human bronchial epithelial cells (BEAS-2B). Metabolomics were used to systematically disclose metabolic characteristics in arsenic-transformed malignant (As-TM) cells. Significantly changed metabolites were enriched in alanine, aspartate and glutamate metabolism, arginine biosynthesis, glutamine and glutamate metabolism, glutathione metabolism, butanoate metabolism, TCA cycle, and arginine and proline metabolism. It is worth noting that glutamate located at the intersection of the enriched metabolism pathways. Glutamine deprivation attenuated the oncogenic phenotypes, including capacity of wound healing and proliferation, in As-TM cells. And the expression levels of mRNA and proteins associated with glutamine metabolism-related transporters and enzymes, including SLC7A11, GCLM, and GCLC, were significantly increased, with SLC7A11 exhibiting the most substantial increase. Moreover, arsenite transformation progressively elevated SLC7A11 mRNA and protein levels over time. The SLC7A11 inhibitor sulfasalazine remarkably attenuated arsenite-induced oncogenic phenotypes. Collectively, our data suggest that chronic arsenite exposure enhances glutamine metabolism through upregulation of SLC7A11, thereby promoting cell proliferation and malignant transformation. These results provide new insights for preventive and therapeutic strategies for lung cancer linked to arsenic exposure.
Collapse
Affiliation(s)
- Xin Fang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yihui Ruan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Xianhang Yin
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Junyi Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Chen Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yuxin Hu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Huihui Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, 110122, China
| | - Jingbo Pi
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yuanyuan Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
46
|
Wang W, Chaudhary R, Szpendyk J, El Khalki L, Yousafzai NA, Chan R, Desai A, Sossey-Alaoui K. Kindlin-2-Mediated Hematopoiesis Remodeling Regulates Triple-Negative Breast Cancer Immune Evasion. Mol Cancer Res 2025; 23:450-462. [PMID: 39918417 DOI: 10.1158/1541-7786.mcr-24-0698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/09/2024] [Accepted: 02/05/2025] [Indexed: 05/03/2025]
Abstract
Triple-negative breast cancer (TNBC) presents significant clinical challenges because of its limited treatment options and aggressive behavior, often associated with poor prognosis. This study focuses on kindlin-2, an adapter protein, and its role in TNBC progression, particularly in hematopoiesis-mediated immune evasion. TNBC tumors expressing high levels of kindlin-2 induce a notable reshaping of hematopoiesis, promoting the expansion of myeloid cells in the bone marrow and spleen. This shift correlated with increased levels of neutrophils and monocytes in tumor-bearing mice over time. Conversely, genetic knockout (KO) of kindlin-2 mitigated this myeloid bias and fostered T-cell infiltration within the tumor microenvironment, indicating the pivotal role of kindlin-2 in immune modulation. Further investigations revealed that kindlin-2 deficiency led to reduced expression of PD-L1, a critical immune checkpoint inhibitor, in TNBC tumors. This molecular change sensitized kindlin-2-deficient tumors to host antitumor immune responses, resulting in enhanced tumor suppression in immunocompetent mouse models. Single-cell RNA sequencing, bulk RNA sequencing, and IHC data supported these findings by highlighting enriched immune-related pathways and increased infiltration of immune cells in kindlin-2-deficient tumors. Therapeutically, targeting PD-L1 in kindlin-2-expressing TNBC tumors effectively inhibited tumor growth, akin to the effects observed with genetic kindlin-2 KO or PD-L1 KO. Our data underscore kindlin-2 as a promising therapeutic target in combination with immune checkpoint blockade to bolster antitumor immunity and counteract resistance mechanisms typical of TNBC and other immune-evasive solid tumors. Implications: Kindlin-2 regulates tumor immune evasion through the systemic modulation of hematopoiesis and PD-L1 expression, which warrants therapeutic targeting of kindlin-2 in patients with TNBC.
Collapse
Affiliation(s)
- Wei Wang
- MetroHealth System, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Lamyae El Khalki
- Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Neelum Aziz Yousafzai
- Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Ricky Chan
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Amar Desai
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Khalid Sossey-Alaoui
- MetroHealth System, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| |
Collapse
|
47
|
Jordan MR, Mendoza-Munoz PL, Pawelczak KS, Turchi JJ. Targeting DNA damage sensors for cancer therapy. DNA Repair (Amst) 2025; 149:103841. [PMID: 40339280 DOI: 10.1016/j.dnarep.2025.103841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/18/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
DNA damage occurs from both endogenous and exogenous sources and DNA damaging agents are a mainstay in cancer therapeutics. DNA damage sensors (DDS) are proteins that recognize and bind to unique DNA structures that arise from direct DNA damage or replication stress and are the first step in the DNA damage response (DDR). DNA damage sensors are responsible for recruiting transducer proteins that signal downstream DNA repair pathways. As the initiating proteins, DDS are excellent candidates for anti-cancer drug targeting to limit DDR activation. Here, we review four major DDS: PARP1, RPA, Ku, and the MRN complex. We briefly describe the cellular DDS functions before analyzing the structural mechanisms of DNA damage sensing. Lastly, we examine the current state of the field towards inhibiting each DDS for anti-cancer therapeutics and broadly discuss the therapeutic potential for DDS targeting.
Collapse
Affiliation(s)
- Matthew R Jordan
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Pamela L Mendoza-Munoz
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - John J Turchi
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States; NERx BioSciences, Indianapolis, IN, United States.
| |
Collapse
|
48
|
Sugár SN, Molnár BA, Bugyi F, Kecskeméti G, Szabó Z, Laczó I, Harkó T, Moldvay J, Turiák L. Glycoproteomics Analysis of Triple Wild-Type Lung Adenocarcinoma Tissue Samples. J Proteome Res 2025; 24:2419-2429. [PMID: 40175289 PMCID: PMC12053933 DOI: 10.1021/acs.jproteome.4c01063] [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: 11/27/2024] [Revised: 02/21/2025] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
Lung cancer has both high incidence and mortality, making it the leading cause of cancer-related mortality worldwide. It is a highly heterogeneous disease, with several histological subtypes and genetic alterations that influence prognosis and available treatment options. Here, we focus on the triple wild-type (TWT) subtype of lung adenocarcinoma (LUAD) that lacks the three most common actionable genetic alterations, subsequently making targeted therapies inaccessible. In this study, our aim was the mass spectrometry-based proteomic and N-glycoproteomic characterization of tumor and adjacent normal lung tissue regions from individuals (n = 12) with TWT LUAD. We found several proteins previously identified as potential prognostic or diagnostic biomarkers in LUAD and described dysregulated biological processes, giving an overview of the general differences between healthy and tumor tissue. Also, we highlight specific signatures detected using N-glycoproteomics and discuss their potential and importance based on data from databases and literature. To the best of our knowledge, this is the first N-glycoproteomics-focused study on TWT LUAD, and it could provide a valuable resource for further studies into this less well characterized subtype of lung cancer. For instance, we report altered N-glycosylation for several glycoproteins implicated in LUAD and other cancers that could have functional importance connected to the disease.
Collapse
Affiliation(s)
- Simon Nándor Sugár
- MTA-HUN-REN
TTK Lendület (Momentum) Glycan Biomarker Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja
2, Budapest H-1117, Hungary
| | - Balázs András Molnár
- MTA-HUN-REN
TTK Lendület (Momentum) Glycan Biomarker Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja
2, Budapest H-1117, Hungary
| | - Fanni Bugyi
- MTA-HUN-REN
TTK Lendület (Momentum) Glycan Biomarker Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja
2, Budapest H-1117, Hungary
- Hevesy
György PhD School of Chemistry, ELTE
Eötvös Loránd University, Pázmány Péter Sétány
1/A, Budapest H-1117, Hungary
| | - Gábor Kecskeméti
- Department
of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm Square 8, Szeged H-6720, Hungary
| | - Zoltán Szabó
- Department
of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm Square 8, Szeged H-6720, Hungary
| | - Ibolya Laczó
- Békés
County Central Hospital, Semmelweis Utca 1, Gyula, H-5700, Hungary
| | - Tünde Harkó
- National
Korányi Institute of Pulmonology, Korányi Frigyes Street 1, Budapest, H-1121, Hungary
| | - Judit Moldvay
- National
Korányi Institute of Pulmonology, Korányi Frigyes Street 1, Budapest, H-1121, Hungary
- Pulmonology
Clinic, Albert Szent-Györgyi Medical School, University of Szeged, Alkotmány Street 36, Deszk H-6771, Hungary
| | - Lilla Turiák
- MTA-HUN-REN
TTK Lendület (Momentum) Glycan Biomarker Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja
2, Budapest H-1117, Hungary
| |
Collapse
|
49
|
Zhang X, Fam KT, Dai T, Hang HC. Microbiota mechanisms in cancer progression and therapy. Cell Chem Biol 2025:S2451-9456(25)00128-X. [PMID: 40334660 DOI: 10.1016/j.chembiol.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025]
Abstract
The composition of the microbiota in patients has been shown to correlate with cancer progression and response to therapy, highlighting unique opportunities to improve patient outcomes. In this review, we discuss the challenges and advancements in understanding the chemical mechanisms of specific microbiota species, pathways, and molecules involved in cancer progression and treatment. We also describe the modulation of cancer and immunotherapy by the microbiota, along with approaches for investigating microbiota enzymes and metabolites. Elucidating these specific microbiota mechanisms and molecules should offer new opportunities for developing enhanced diagnostics and therapeutics to improve outcomes for cancer patients. Nonetheless, many microbiota mechanisms remain to be determined and require innovative chemical genetic approaches.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Kyong Tkhe Fam
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Tingting Dai
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA; Department of Chemistry, Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
50
|
Lin Y, Zhang Y, Huang T, Chen J, Li G, Zhang B, Xu L, Wang K, He H, Chen H, Liu D, Guo S, He X, Lan P. Arginine Deprivation Induces Quiescence and Confers Vulnerability to Ferroptosis in Colorectal Cancer. Cancer Res 2025; 85:1663-1679. [PMID: 39992728 PMCID: PMC12046318 DOI: 10.1158/0008-5472.can-24-1940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 11/21/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
Metabolic reprogramming is a hallmark of cancer. Rewiring of amino acid metabolic processes provides the basis for amino acid deprivation therapies. In this study, we found that arginine biosynthesis is limited in colorectal cancer because of the deficiency of ornithine transcarbamylase. Accordingly, colorectal cancer cells met the demand for arginine by increasing external uptake. The addiction to environmental arginine resulted in the susceptibility of colorectal cancer to arginine deprivation, which dramatically decreased proliferation in colorectal cancer cells and promoted these cells to enter a reversible quiescence state. Arginine deprivation induced quiescence by activating the AMPK-p53-p21 pathway. RNA sequencing data indicated that colorectal cancer cells may be vulnerable to ferroptosis during arginine deprivation and the combination of ferroptosis inducers and arginine deprivation strongly impeded tumor growth in vivo. These findings suggest that dietary modification combined with ferroptosis induction could be a potential therapeutic strategy for colorectal cancer. Significance: Colorectal cancer dependency on arginine uptake creates a metabolic vulnerability to arginine deficiency that causes cell cycle arrest and ferroptosis sensitivity, highlighting arginine deprivation plus ferroptosis induction as a promising treatment.
Collapse
Affiliation(s)
- Yanyun Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanhong Zhang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianze Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junguo Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guanman Li
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bin Zhang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Xu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Anaesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Danling Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuang Guo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaosheng He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping Lan
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| |
Collapse
|