1
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [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: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
2
|
Yuan Q, Li X, Chen X, Xiao J, Zhang J. 7‑Difluoromethoxyl‑5,4'‑di‑n‑octylygenistein targets the STAT3 pathway by upregulating microRNA‑152‑3p expression to inhibit self‑renewal and tumor growth in non‑small cell lung carcinoma. Oncol Rep 2025; 53:66. [PMID: 40242966 PMCID: PMC12046948 DOI: 10.3892/or.2025.8899] [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: 10/27/2024] [Accepted: 02/25/2025] [Indexed: 04/18/2025] Open
Abstract
MicroRNAs (miRs) serve a pivotal role in the regulation of non‑small cell lung carcinoma (NSCLC). The present study aimed to investigate the antitumor effects of 7‑difluoromethoxyl‑5,4'‑di‑n‑octylygenistein (DFOG), a novel synthetic genistein derivative, on NSCLC, and to elucidate its molecular mechanism. The research focused on whether DFOG inhibited self‑renewal and tumor growth in NSCLC by modulating the miR‑152‑3p/STAT3 signaling pathway. Reverse transcription‑quantitative PCR and western blot analyses were employed to assess miR‑152‑3p expression and phosphorylated‑STAT3 (p‑STAT3) levels. The effects of DFOG on self‑renewal and tumor growth were evaluated via sphere formation and clonogenic assays. Additionally, sphere‑forming cells (SFCs) were enriched using a suspension culture method, and western blot analysis was conducted to examine stemness markers (CD133, CD44, Oct4 and Sox2). The results demonstrated that DFOG inhibited self‑renewal and tumor growth in NSCLC. This effect was associated with increased miR‑152‑3p expression, decreased STAT3 mRNA levels and reduced p‑STAT3 levels in NSCLC cells. Furthermore, inhibition or overexpression of STAT3 did not alter miR‑152‑3p expression but modulated the inhibitory effects of DFOG on self‑renewal and tumor growth. These findings highlighted that DFOG suppressed self‑renewal and tumor growth in SFCs derived from NSCLC by directly targeting STAT3 through the upregulation of miR‑152‑3p.
Collapse
Affiliation(s)
- Qing Yuan
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology and Research Center for Translational Medicine in Colleges and Universities, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Xiang Li
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Xuemei Chen
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jianhui Xiao
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology and Research Center for Translational Medicine in Colleges and Universities, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jiansong Zhang
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
3
|
Vanan AG, Vesal S, Seraj P, Ghezel MA, Eini P, Talebileili M, Asgari Z, Tahmasebi S, Hashemi M, Taheriazam A. DCLK1 in gastrointestinal cancer: A driver of tumor progression and a promising therapeutic target. Int J Cancer 2025; 156:2068-2086. [PMID: 40056091 DOI: 10.1002/ijc.35365] [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: 10/24/2024] [Revised: 01/12/2025] [Accepted: 01/29/2025] [Indexed: 04/05/2025]
Abstract
Cancers of the gastrointestinal (GI) tract, including colorectal, pancreatic, and hepatocellular carcinomas, represent a significant global health burden due to their high incidence and mortality rates. Doublecortin-like kinase 1 (DCLK1), initially identified for its role in neurogenesis, has emerged as a crucial player in GI cancer progression. This review comprehensively examines the multifaceted roles of DCLK1 in GI cancers, focusing on its structural isoforms, functions in normal and inflammatory states, and contributions to cancer progression and metastasis. DCLK1 is overexpressed in various GI cancers and is associated with poor prognosis, enhanced tumorigenic potential, and increased metastatic capacity. The review discusses the molecular mechanisms through which DCLK1 influences cancer stem cell maintenance, epithelial-mesenchymal transition (EMT), and cell survival pathways, as well as its interactions with key signaling pathways such as Notch, WNT/β-catenin, and NF-κB. The potential of DCLK1 as a therapeutic target is also explored, highlighting preclinical and early clinical efforts to inhibit its function using small molecule inhibitors or monoclonal antibodies. Despite significant advancements, further research is needed to fully elucidate DCLK1's role in GI cancers and to develop effective therapeutic strategies targeting this protein.
Collapse
Affiliation(s)
- Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheil Vesal
- Department of Molecular Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Parmida Seraj
- Department of Medicine, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | | | - Pooya Eini
- Toxicological Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Talebileili
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Zeynab Asgari
- Department of Immunology, School of Medicine Kerman University of Medical Sciences, Kerman, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Faculty of Advanced Science and Technology, Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Faculty of Medicine, Department of Orthopedics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
4
|
Qi X, Zhou J, Wang P, Li Y, Li H, Miao Y, Ma X, Luo X, Zhang Z, He Y, Shen W, Zhao W, Cui R, Li C, Zhu H, Lyu J. KLF7-regulated ITGA2 as a therapeutic target for inhibiting oral cancer stem cells. Cell Death Dis 2025; 16:354. [PMID: 40316546 PMCID: PMC12048542 DOI: 10.1038/s41419-025-07689-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
Cancer stem cells (CSCs) play crucial roles in tumor metastasis, therapy resistance, and immune evasion. Identifying and understanding the factors that regulate the stemness of tumor cells presents promising opportunities for developing effective therapeutic strategies. In this study on oral squamous cell carcinoma (OSCC), we confirmed the key role of KLF7 in maintaining the stemness of OSCC. Using chromatin immunoprecipitation sequencing and dual-luciferase assays, we identified ITGA2, a membrane receptor, as a key downstream gene regulated by KLF7 in the maintenance of stemness. Tumor sphere formation assays, flow cytometry analyses, and in vivo limiting dilution tumorigenicity evaluations demonstrated that knocking down ITGA2 significantly impaired stemness. Upon binding to its extracellular matrix (ECM) ligand, type I collagen, ITGA2 activates stemness-associated signaling pathways, including PI3K-AKT, MAPK, and Hippo. TC-I 15, a small-molecule inhibitor of the ITGA2-collagen interaction, significantly sensitizes oral squamous cell carcinoma (OSCC) to cisplatin in xenograft models. In summary, we reveal that the KLF7/ITGA2 axis is a crucial modulator of stemness in OSCC. Our findings suggest that ITGA2 is a promising therapeutic target, offering a novel anti-CSC strategy.
Collapse
Affiliation(s)
- Xin Qi
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Jiang Zhou
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Pan Wang
- Department of Stomatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yunyan Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Haoran Li
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Yuwen Miao
- Zhejiang University, School of Medicine, Affiliated Stomatology Hospital, Hangzhou, Zhejiang, P. R. China
| | - XiaoQing Ma
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiayan Luo
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Zhiling Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Yanling He
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Wenyi Shen
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Wenquan Zhao
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Rutao Cui
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Cang Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China.
| | - Huiyong Zhu
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China.
| | - Jiong Lyu
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China.
| |
Collapse
|
5
|
Yang C, Li Y, Wang Z, Shan H, Zhang G, Meng X, Wang G, Hou Z, Zhao X, Zhang X, Liu A, Bing Y, Lei G, Jin Y, Luo J, Guo L, Yin Y. Identification of a cancer stem cell-like subpopulation that promotes HCC metastasis. JHEP Rep 2025; 7:101302. [PMID: 40242316 PMCID: PMC11999271 DOI: 10.1016/j.jhepr.2024.101302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 04/18/2025] Open
Abstract
Background & Aims Cancer stem cells (CSCs) are well-established drivers of tumorigenesis, but their role in regulating tumor metastasis remains poorly understood. Here, we report the identification and characterization of a cluster of metastasis-promoting CSC-like cells in hepatocellular carcinoma (HCC). Methods CSC-like cells in HCC were identified through the analysis of single cell RNA-sequencing data from 19 HCC samples. The stemness and invasive characteristics of these cells were evaluated using bioinformatical analyses of nine clinical cohorts and experimental validations. Spatial transcriptomics sequencing of 12 HCC samples revealed the cellular interactions between the CSC-like cells and tumor microenvironments, which were validated through gene co-expression analyses and immunohistochemistry. Finally, signaling pathway blockade was used to assess the potential clinical application of CSC-like cells. Results Through comprehensive analyses of single cell RNA-sequencing data from 19 patients with HCC and spatial transcriptomics data from 12 patients with HCC, a metastasis-promoting CSC-like subpopulation was identified. These CSC-like cells expressed high levels of epithelial-mesenchymal transition genes and were associated with poor prognosis of HCC. Histologically, CSC-like cells were enriched in highly aggressive tumors, especially in intrahepatic disseminated foci, where they interacted with immune cells. Functionally, CSC-like cells induced macrophage M2 polarization and T cell exhaustion through the ICAM1 signaling pathway, forming immunosuppressive microenvironments. Downregulation of ICAM1 expression in CSC-like cells suppressed macrophage M2-polarization and T cell exhaustion, thereby reversing antitumor immune effects. Conclusions Our study identified a metastasis-promoting CSC subpopulation, providing a potential perspective for CSC-targeted therapies in HCC. Impact and implications The heterogeneity of CSCs in HCC has been identified, yet the identification and characterization of metastasis-promoting CSC subpopulations remain unexplored. Here, we identified a CSC-like tumor cell subpopulation that promotes HCC metastasis by increasing cell invasiveness and suppressing antitumor immune responses via the ICAM1 signaling pathway. Our study uncovers novel mechanisms of HCC metastasis from the perspective of CSCs, and proposes potential tumor therapeutic strategies by inhibiting cellular interactions between CSC-like cells and immune cells.
Collapse
Affiliation(s)
- Chunyuan Yang
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yang Li
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhaohai Wang
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Hui Shan
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Guangze Zhang
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xiangyan Meng
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Guangxi Wang
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Zhiyuan Hou
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xuyang Zhao
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xin Zhang
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Anhang Liu
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yuntao Bing
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Guanglin Lei
- Senior Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, 100039 Beijing, China
| | - Yan Jin
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jianyuan Luo
- Department of Medical Genetics, School of Basic Medical Sciences Peking University, Beijing 100191, China
| | - Limei Guo
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
6
|
Guo Q, Qin H, Chen Z, Zhang W, Zheng L, Qin T. Key roles of ubiquitination in regulating critical regulators of cancer stem cell functionality. Genes Dis 2025; 12:101311. [PMID: 40034124 PMCID: PMC11875185 DOI: 10.1016/j.gendis.2024.101311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/23/2024] [Accepted: 03/07/2024] [Indexed: 03/05/2025] Open
Abstract
The ubiquitin (Ub) system, a ubiquitous presence across eukaryotes, plays a crucial role in the precise orchestration of diverse cellular protein processes. From steering cellular signaling pathways and orchestrating cell cycle progression to guiding receptor trafficking and modulating immune responses, this process plays a crucial role in regulating various biological functions. The dysregulation of Ub-mediated signaling pathways in prevalent cancers ushers in a spectrum of clinical outcomes ranging from tumorigenesis and metastasis to recurrence and drug resistance. Ubiquitination, a linchpin process mediated by Ub, assumes a central mantle in molding cellular signaling dynamics. It navigates transitions in biological cues and ultimately shapes the destiny of proteins. Recent years have witnessed an upsurge in the momentum surrounding the development of protein-based therapeutics aimed at targeting the Ub system under the sway of cancer stem cells. The article provides a comprehensive overview of the ongoing in-depth discussions regarding the regulation of the Ub system and its impact on the development of cancer stem cells. Amidst the tapestry of insights, the article delves into the expansive roles of E3 Ub ligases, deubiquitinases, and transcription factors entwined with cancer stem cells. Furthermore, the spotlight turns to the interplay with pivotal signaling pathways the Notch, Hedgehog, Wnt/β-catenin, and Hippo-YAP signaling pathways all play crucial roles in the regulation of cancer stem cells followed by the specific modulation of Ub-proteasome.
Collapse
Affiliation(s)
- Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, Guizhou 550014, China
| | - Zelong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Artificial Intelligence and IoT Smart Medical Engineering Research Center of Henan Province, Zhengzhou, Henan 450008, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Tingting Qin
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| |
Collapse
|
7
|
Zhao Y, Lu L, Chen X, Yin Q. Natural compounds targeting ferroptosis in ovarian cancer: Research progress and application potential. Pharmacol Res 2025; 215:107729. [PMID: 40194611 DOI: 10.1016/j.phrs.2025.107729] [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: 11/14/2024] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
Ovarian cancer (OC) is among the most common malignancies in the female reproductive system, marked by high rates of recurrence and mortality. Conventional chemotherapy, however, faces limitations due to the development of drug resistance, which hinders its effectiveness. Ferroptosis, an atypical form of programmed cell death distinct from autophagy, apoptosis, and necrosis, the relationship with tumors has become a hot research area in cancer studies in recent years. Anticancer therapies that target ferroptosis show strong potential in improving prognosis and counteracting chemotherapy resistance. Natural compounds, as a valuable source of novel targeted anticancer agents, its significant role in inhibiting tumor cell proliferation and metastasis and improving therapeutic sensitivity has been demonstrated in numerous existing studies. This review summarizes a range of natural compounds that target ferroptosis in OC cells, discussing their active components, mechanisms of action, and therapeutic potential, thereby providing useful insights for future targeted therapy and research in OC.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.
| | - Lichao Lu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China.
| | - Xingying Chen
- Yuebei People's Hospital, Shaoguan, Guangdong 512000, China.
| | - Qiaozhi Yin
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.
| |
Collapse
|
8
|
Liu Q, Wang Y, Song M, Huang J, Shi J, Sun W, Ji X, Chang Y, Ma B, Zhang P, Yan Y, Zhang H. CCL20/CXCL5 Drives Crosstalk Between Anaplastic Thyroid Cancer Stem Cells and Tumor-Associated Macrophages to Promote Tumor Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405399. [PMID: 40091357 PMCID: PMC12061268 DOI: 10.1002/advs.202405399] [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] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 02/26/2025] [Indexed: 03/19/2025]
Abstract
The dynamic interplay between tumor-associated macrophages (TAMs) and anaplastic thyroid cancer (ATC) shapes the tumor microenvironment and facilitates ATC progression. However, the mechanisms of communication between TAMs and anaplastic thyroid cancer stem cells (ATCSCs) remain largely unelucidated. Integrative analyses of single-cell RNA sequencing, cytokine/chemokine arrays, proteomics, and mRNA expression datasets are performed to reveal crosstalk between TAMs and ATCSCs and signaling pathways in ATCSCs. Subsequently, in vitro experiments are performed to validate the regulatory effects of key cytokines on ATCSC stemness. Last, xenogeneic orthotopic thyroid ATCSCs transplantation models are utilized to corroborate the regulatory effect of cytokines on stemness. CCL20 derived from THP-1-M2 activates the IRAK-1/NF-κB1/2 signaling pathway in ATCSCs, thereby positively regulating stemness characteristics and upregulating CXCL5 secretion. ATCSCs not only exhibit autocrine CXCL5 participation in the regulation of stemness but also demonstrate paracrine CXCL5 activity to recruit THP-1-Mφ and maintain the M2 phenotype. CCL20 and CXCL5 are involved in the crosstalk between TAMs and ATCSCs. The CCL20/CXCL5 axis plays a crucial role in the interaction between TAMs and ATCSCs, establishing a progressive tumor microenvironment.
Collapse
Affiliation(s)
- Qi Liu
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Yan Wang
- Department of PharmacologySchool of PharmacyChina Medical UniversityShenyang110122P. R. China
| | - Mingyuan Song
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Jiapeng Huang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Jinyuan Shi
- Department of Thyroid SurgeryGeneral SurgeryQilu Hospital of Shandong UniversityJinan250012P. R. China
| | - Wei Sun
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Xiaoyu Ji
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Yuang Chang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Bing Ma
- Department of Clinical Epidemiology and Evidence‐based MedicineThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Ping Zhang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Yuanyuan Yan
- Department of PharmacologySchool of PharmacyChina Medical UniversityShenyang110122P. R. China
| | - Hao Zhang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| |
Collapse
|
9
|
Mai W, Tang Y, He W, Zhu C, Feng B, Lyu J, Chen Z. Construction and Evaluation of a Prognostic Columnar Graphic Model for Adult Patients with Diffuse Midline Gliomas. World Neurosurg 2025; 197:123901. [PMID: 40090411 DOI: 10.1016/j.wneu.2025.123901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVE To explore the prognostic factors of adult patients with diffuse midline glioma (DMG), and to further construct and evaluate prognostic columnar graphic models to provide some reference for the clinical management of this group of patients. METHODS We included adult patients with histologically confirmed DMG from the SEER (Surveillance Epidemiology and End Results) database (2004-2015), dividing them into training and validation sets (7:3 ratio). Using Kaplan-Meier and Cox regression analyses, we determined independent prognostic factors for overall survival (OS) and cancer-specific survival (CSS). Prognostic column-line graphic models were developed for OS and CSS, incorporating patient demographics and clinical characteristics. The models underwent internal and external validation, with performance assessed using the Concordance Index, area under the curve values, and calibration plots. RESULTS The study encompassed 226 patients, showing age, tumor extension, and World Health Organization grades as significant prognostic factors. The constructed models for OS and CSS showed moderate reliability and predictive accuracy, with Concordance Index values of 0.786 (OS) and 0.79 (CSS) in the training set and 0.743 (OS) and 0.787 (CSS) in the validation set. Calibration plots and decision curve analysis confirmed the clinical usefulness of the models. CONCLUSIONS The column-line graphic prediction models for OS and CSS have moderately reliable predictive efficacy and help clinicians to better assess the prognosis and provide individualized treatment options for adults with DMG.
Collapse
Affiliation(s)
- Wangxiang Mai
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yuting Tang
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Weiyi He
- Department of Pediatrics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Changsen Zhu
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Bing Feng
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, Guangdong, China
| | - Zhuoming Chen
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Lie Q, Jiang H, Lu X, Chen Z, Liang J, Zhang Y, Chao H. Photo-Activated Ferrocene-Iridium(III) Prodrug Induces Immunogenic Cell Death in Melanoma Stem Cells. J Med Chem 2025; 68:8894-8906. [PMID: 40233007 DOI: 10.1021/acs.jmedchem.5c00533] [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: 04/17/2025]
Abstract
Cancer stem cells (CSCs) are key contributors to tumor resistance, recurrence, and metastasis. Conventional chemotherapy often fails to target and eradicate CSCs, significantly impairing their therapeutic efficacy. Herein, we design and synthesize a photoactivated ferrocene-iridium(III) complex (Ir-3) to achieve immunotherapy against melanoma cells (including stem cells). In short, Ir-3 effectively targets mitochondria and dissociates under light irradiation to produce a cytotoxic Ir(III) photosensitizer and Fe2+ ions. They can generate reactive oxygen species by the Fenton reaction, robustly induce ferroptosis and autophagy, and eventually trigger immunogenic cell death in melanoma cells (including stem cells). Furthermore, under light exposure, Ir-3 effectively inhibits stem cell-related properties and promotes macrophage-mediated phagocytosis of melanoma stem cells. For in vivo studies, Ir-3 is encapsulated in DSPE-PEG 2000 to form tumor-targeting Ir-3@PEG nanoparticles. After photoactivation, Ir-3@PEG can significantly inhibit primary and distant tumors, effectively inhibit the stemness of melanoma stem cells, and induce innate and adaptive immune responses.
Collapse
Affiliation(s)
- Qiaoshan Lie
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Hui Jiang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Xiangwan Lu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Zhuoli Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Jinzhe Liang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Yan Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510006, P. R. China
- MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 400201, P. R. China
| |
Collapse
|
11
|
Liu XH, Zhong NN, Yi JR, Wang GR, Xiao Y, Zhu ZR, Man QW, Li Z, Liu B, Bu LL. NR2F2 and its contribution to lymph node metastasis in oral squamous cell carcinoma. Cell Signal 2025; 132:111814. [PMID: 40262715 DOI: 10.1016/j.cellsig.2025.111814] [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: 12/12/2024] [Revised: 03/30/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVES To investigate the role of cancer stem cells (CSCs) in lymph node metastasis (LNM) of oral squamous cell carcinoma (OSCC), focusing on the expression and biological significance of nuclear receptor subfamily 2 group F member 2 (NR2F2). METHODS Single-cell RNA sequencing data from OSCC patients were analyzed using the CytoTRACE algorithm to assess stemness. Gene set scores were calculated with the irGSEA and GSVA R packages. GO and KEGG analyses identified enriched pathways. NR2F2 and CD44 expression in OSCC and lymph nodes (LNs) were validated via immunohistochemistry and immunofluorescence. NR2F2/Nr2f2 overexpression and knockdown cell lines were established, with stemness markers confirmed by Western blot. Functional assays evaluated stemness, proliferation, migration, and invasion capabilities of OSCC cells. In vivo experiments evaluated the ability of NR2F2 to promote tumor growth and metastasis. Bulk RNA sequencing and drug sensitivity analyses explored NR2F2-related mechanisms and drug responses. RESULTS CSCs in OSCC were divided into five subgroups, with NR2F2 identified as the key gene in CSC4, the subgroup with the highest stemness, and found to be overexpressed in metastatic LNs. Immunohistochemistry showed NR2F2 overexpression in OSCC, associated with LNM. Immunofluorescence confirmed co-expression of NR2F2 and CD44 in metastatic OSCC and LNs. Overexpression of NR2F2 enhanced stemness, proliferation, and migration of OSCC cells. In vivo experiments showed that NR2F2 promoted the growth and LNM of OSCC. Bulk RNA sequencing revealed that NR2F2 is involved in multiple pathways and plays a role in LNM. Trametinib was identified as a sensitive drug. CONCLUSION NR2F2 is associated with the maintenance of tumor stemness and may influence LNM in OSCC by promoting tumor cell proliferation and migration.
Collapse
Affiliation(s)
- Xuan-Hao Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Jing-Rui Yi
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Zheng-Rui Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Qi-Wen Man
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; Department of Oral & Maxillofacial - Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Zheng Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan 430071, China.
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; Department of Oral & Maxillofacial - Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430072, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; Department of Oral & Maxillofacial - Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
12
|
Fu J, Liu F, Bai S, Jiang X, Song H, Zhang M, Zhao R, Ouyang T, Yu M, Qian H, Xu S, Huo Y, Yang X, Chen L, Cao D, Guo T, Wei Y, Li L, Wang H. Circular RNA CDYL facilitates hepatocellular carcinoma stemness and PD-L1 + exosomes-mediated immunotherapy resistance via stabilizing hornerin protein by blocking synoviolin 1-mediated ubiquitination. Int J Biol Macromol 2025; 310:143246. [PMID: 40250664 DOI: 10.1016/j.ijbiomac.2025.143246] [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: 01/17/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Despite the revolutionary progress in cancer immunotherapy, only a minority of hepatocellular carcinoma (HCC) patients respond to immune checkpoint inhibitors (ICIs). In this study, we found that the oncogenic circular RNA Circ-CDYL in HCC influences the efficacy of immunotherapy and the stemness characteristics of HCC cells by interacting with the hornerin (HRNR) protein. The degraded anti-PD-L1 immunotherapy responses induced by Circ-CDYL and HRNR were confirmed by peripheral blood mononuclear cells (PBMCs) killing assays in HCC cells, patient-derived organoids, and humanized immune system mouse models. Furthermore, Circ-CDYL interference reversed the cytotoxicity and proliferation of CD8+ T cells, resulting in ameliorated immune evasion in tumor spheroids upon anti-PD-L1 treatment. Mechanistically, Circ-CDYL upregulated HRNR expression by stabilizing the HRNR protein through the prevention of its degradation by the E3 ubiquitin ligase synoviolin 1 (SYVN1), which in sequence promoted the phosphorylation of the mTORC1 and p70S6K substrate. The abnormally activated mTORC1-p70S6K signaling increases the stemness of HCC cells and upregulates PD-L1 expression, which may attenuate anti-PD-L1 therapy efficacy via PD-L1+ exosomes. Our study revealed the mechanism by which Circ-CDYL and HRNR regulate the sensitivity of HCC to anti-PD-L1 therapy, and the findings have potential implications for patient stratification and clinical decision-making in HCC immunotherapy.
Collapse
Affiliation(s)
- Jingbo Fu
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Fuyan Liu
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Shilei Bai
- Department of Hepatic Surgery II, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Xue Jiang
- National Center for Liver Cancer, Shanghai, China
| | - Hao Song
- International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Man Zhang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Ru Zhao
- International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Tao Ouyang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Miao Yu
- National Center for Liver Cancer, Shanghai, China
| | - Haihua Qian
- National Center for Liver Cancer, Shanghai, China
| | - Shuo Xu
- International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| | - Yunfei Huo
- National Center for Liver Cancer, Shanghai, China
| | - Xinwei Yang
- Department of Biliary Tract Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Lu Chen
- National Center for Liver Cancer, Shanghai, China
| | - Dan Cao
- International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Tao Guo
- Department of integrated traditional Chinese and Western Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yanping Wei
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China.
| | - Liang Li
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China.
| | - Hongyang Wang
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepato-biliary Surgery Institute, Second Military Medical University, Shanghai, China; National Center for Liver Cancer, Shanghai, China
| |
Collapse
|
13
|
Zhang J, Dong C, Chen Z, Hua R, Li Z, Lin Y, Wang Y, Feng T, Dai J. Hedgehog pathway inhibitor HhAntag suppresses virus infection via the GLI-S1PR axis. Cell Signal 2025; 132:111807. [PMID: 40239727 DOI: 10.1016/j.cellsig.2025.111807] [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: 12/15/2024] [Revised: 03/23/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
The interplay between various signaling pathways, including tumor development, immune response, and viral infection, suggests potential mutual regulation within biological systems. To explore this, we screened 85 inhibitors targeting the Notch, Hedgehog, and Wnt signaling pathways to identify the potential antiviral candidates. Using two reporter viruses (VSV-GFP and DENV-Luc), we identified novel inhibitors with antiviral properties. Notably, the Hedgehog pathway inhibitor HhAntag exhibited broad-spectrum antiviral activity, significantly reducing the replication of viruses such as VSV, DENV, ZIKV, and SFTSV. The inhibitory effects of HhAntag were consistent with the downregulation of its target protein, GLI1; while overexpression of GLI1 promoted viral infection. HhAntag did not interfere with viral attachment, entry, or early transcription but specifically inhibited viral protein translation. Additionally, RNA-seq analysis revealed reduced expression of sphingosine-1-phosphate (S1P) signaling pathway receptors, S1PR1 and S1PR5, following HhAntag treatment. HhAntag suppresses virus infection via the GLI-S1PR axis. This study revealed the interplay between tumor-associated Hedgehog (Hh) pathway and viral infection and highlights the potential of HhAntag as a broad-spectrum antiviral drug.
Collapse
Affiliation(s)
- Jinyu Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Children's Hospital of Soochow University, Institute of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China; Central Laboratory, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunsheng Dong
- Jiangsu Key Laboratory of Infection and Immunity, Children's Hospital of Soochow University, Institute of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Zhiqiang Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Runbin Hua
- Jiangsu Key Laboratory of Infection and Immunity, Children's Hospital of Soochow University, Institute of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Zhuozheng Li
- School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China
| | - Yuzhuo Lin
- The Second Clinical Medical School of Nanjing Medical University, Nanjing 211166, China
| | - Yuqing Wang
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou 215000, China.
| | - Tingting Feng
- Central Laboratory, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Jianfeng Dai
- Jiangsu Key Laboratory of Infection and Immunity, Children's Hospital of Soochow University, Institute of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China; MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
14
|
Boyacıoğlu Ö, Kalali BD, Tongün E, Korkusuz P. A Niche-Based Perspective to Stem and Cancer Stem Cells of the Lung. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40178798 DOI: 10.1007/5584_2025_858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Lungs carry the principle function for the conduction and exchange of air through the primary, secondary, tertiary bronchi, bronchioles, and alveoli, resulting in the exchange of oxygen to carbon dioxide within the human tissues. Lung stem and progenitor cells enable differentiation of parenchymal and stromal elements and provide homeostasis and regeneration in the microenvironment against pulmonary diseases. Tumor-initiating cancer cells (TICs) refer to a subpopulation named as cancer stem cells (CSCs) of lung cancer exhibiting high self-renewal and proliferation capacity by Notch, Hippo, Hedgehog, and Wnt signaling pathways that leads to tumor development or recurrence. Lung cancer stem cells (LCSCs) are characterized by distinct genotypic or phenotypic alterations compared to healthy lung stem cells (LSCs) that provide a potential target to treat lung cancer. Therefore, understanding the cascades responsible for the transformation of healthy to CSCs is essential to develop new targeted therapy approaches. In this chapter, we precisely highlight the latest researches on LSCs and CSCs, key signaling mechanisms within the perspective of novel targeted therapy strategies.
Collapse
Affiliation(s)
- Özge Boyacıoğlu
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, Turkey
- Department of Medical Biochemistry, Faculty of Medicine, Atılım University, Gölbaşı, Ankara, Turkey
| | - Berfin Deniz Kalali
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, Turkey
| | - Ege Tongün
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Sıhhiye, Ankara, Turkey.
- METU MEMS Center, Ankara, Turkey.
| |
Collapse
|
15
|
Yang C, Zhang Y, Yan L, Liu A, Li F, Li Y, Zhang Y. Comprehensive Analysis of GPSM2: From Pan-Cancer Analysis to Experimental Validation. J Cell Mol Med 2025; 29:e70527. [PMID: 40208185 PMCID: PMC11984320 DOI: 10.1111/jcmm.70527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/04/2024] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
G-protein signalling modulator 2 (GPSM2) plays an important role in maintaining cell polarisation and regulating the cell cycle; however, a systematic and comprehensive analysis of GPSM2 in cancer is still lacking. Using extensive multi-omics data, we explored the pan-cancer expression levels of GPSM2 from multiple perspectives and its association with prognosis, diagnosis, tumour stemness, immune-related genes, immune cell infiltration, genomic instability, and response to immunotherapy. We also elucidated the potential pan-cancer biological functions of GPSM2 using gene set enrichment analysis (GSEA) and searched for targeted drugs that affect GPSM2 expression using connectivity map analysis. To elucidate the effect of GPSM2 on colon cancer, we evaluated its effect on the biological behaviour of two colon cancer cell lines. In this study, GPSM2 was systematically analysed and shown to have satisfactory performance in disease diagnosis and prognostic prediction of various cancers. G-protein signalling modulator 2 plays an important role in the genesis and development of various tumours and is a potential tumour diagnostic and prognostic biomarker as well as an anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Chunjiao Yang
- Department of OncologyThe Fifth Affiliated Hospital of Guangxi Medical University & The First People's Hospital of NanningNanningChina
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Yuzhe Zhang
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Lirong Yan
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Aoran Liu
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Fang Li
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Yanke Li
- Department of Anorectal SurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Ye Zhang
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
16
|
Gu P, Liu J, Gao T, Ma Q, Gao S, Li N, Zhang W, Xia Z, Yang Q, Mu W, Liang S, Fu S, Yuan S, Wei S, Liu J, Yang Y, Yan X, Liu Y, Wang C, Zhang N. Temperature-Sensitive Nano-GOx Combined with Downregulation of Tumor Stemness to Initiate Robust Antitumor Efficacy. ACS NANO 2025; 19:11738-11755. [PMID: 40105115 DOI: 10.1021/acsnano.4c12962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
GOx-mediated glucose depletion offers an alternative noninvasive strategy for tumor therapy, but its lower catalytic activity in vivo limits its clinical application. Herein, we designed a temperature-sensitive nano-GOx (NG) that was constructed by gold nanorods chemically modified with GOx (AuNRs-GOx) and coated with temperature-sensitive lipids. The chemical linkage could maintain the natural conformation of GOx, ensuring that NG exerted powerful catalytic activity within the tumor and initiated antitumor immune response through moderate starvation and mild photothermal therapy (mPTT) to coregulating dendritic cells (DCs) and tumor-associated macrophages (TAMs). Ulteriorly, VTNG was obtained by NG coloading with verteporfin (VP) and evofosfamide (TH-302). VTNG demonstrated temperature-sensitive triggered drug release when exposed to near-infrared laser irradiation. NG exacerbated the degree of TME hypoxia and facilitated the activation of TH-302. Meanwhile, VP enhanced tumor cell sensitivity by decreasing the stemness of the tumor cells, thus realizing the effective killing of tumor cells and further enhancing the therapeutic effect of NG. Notably, VTNG had a significant antitumor effect in melanoma models compared with first-line melanoma therapy and formed an immune memory effect. In conclusion, VTNG provided an effective approach to enhance the therapeutic effect of GOx for tumor treatment.
Collapse
Affiliation(s)
- Panpan Gu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Jinhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Tong Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Shuying Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Nan Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Weihan Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Zhenxing Xia
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Qinglin Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Shuang Liang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Shijun Yuan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Suyun Wei
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Jie Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Yuxin Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Xiaoxin Yan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Cong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| |
Collapse
|
17
|
Wu J, Liu X, Huang S, Liu W. Identification of a Cancer Stem Cell-Related Gene Signature in Hepatocellular Carcinoma Based on Single-Cell RNA-Seq and Bulk RNA-Seq Analysis. Int J Mol Sci 2025; 26:2933. [PMID: 40243557 PMCID: PMC11988464 DOI: 10.3390/ijms26072933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) are a heterogeneous group of tumor cells that play a significant role in tumorigenesis, therapeutic resistance, and recurrence in liver hepatocellular carcinoma (LIHC). This study combines clinical data sets from The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC) with bulk RNA sequencing data. This study also features the GSE156625 single-cell RNA sequencing (scRNA) data set from the GEO to explore the prognostic significance of CSC biomarkers (BCSCs) in LIHC. In this research, we introduce a developed prognostic risk model that relies on nine specific BCSCs, including ADM, CCL5, CD274, DLGAP5, HOXD9, IGF1, S100A9, SOCS2, and TNFRSF11B. It was found that high-risk patients experience shorter overall survival rates when compared to low-risk patients. Additionally, the study characterized the composition of immune cells within the tumor microenvironment (TME) and revealed significant variations in gene-expression levels and mutation rates between different risk groups. The model suggests that liver cancer progression might be driven by immune evasion independent of PD-L1 and highlights the potential of the low-risk BCSC group being sensitive to various treatments. Our findings offer a promising foundation for personalized LIHC therapy and highlight the need for further experimental validation of the roles of these CSCs in disease progression.
Collapse
Affiliation(s)
- Jing Wu
- Medical School, Hubei Minzu University, Enshi 445000, China; (X.L.); (S.H.)
| | - Xu Liu
- Medical School, Hubei Minzu University, Enshi 445000, China; (X.L.); (S.H.)
| | - Sheng Huang
- Medical School, Hubei Minzu University, Enshi 445000, China; (X.L.); (S.H.)
| | - Wei Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
18
|
Ou Z, Zhu L, Chen X, Liu T, Cheng G, Liu R, Zhang S, Tan W, Lin D, Wu C. Hypoxia-Induced Senescent Fibroblasts Secrete IGF1 to Promote Cancer Stemness in Esophageal Squamous Cell Carcinoma. Cancer Res 2025; 85:1064-1081. [PMID: 39661488 DOI: 10.1158/0008-5472.can-24-1185] [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: 04/12/2024] [Revised: 08/12/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Cancer-associated fibroblasts (CAF) contribute to cancer initiation and progression and play a pivotal role in therapeutic response and patient prognosis. CAFs exhibit functional and phenotypic heterogeneity, highlighting the need to clarify the specific subtypes of CAFs to facilitate the development of targeted therapies against protumorigenic CAFs. In this study, using single-cell RNA sequencing on patient samples of esophageal squamous cell carcinoma (ESCC), we identified a CAF subcluster associated with tumor stemness that was enriched in genes associated with hypoxia and senescence. The CAF subpopulation, termed as hypoxia-induced senescent fibroblasts (hsCAF), displayed high secretion of insulin-like growth factor 1 (IGF1). The hsCAFs inhibited AMP-activated protein kinase (AMPK) activity in cancer cells via IGF1 to promote tumor stemness. The formation of hsCAFs was induced by the synergetic effect of hypoxia and cancer cells. Activation of nuclear factor erythroid 2-related factor 2 (NRF2) in cancer cells under hypoxia drove IL1α production to trigger CAF senescence and IGF1 secretion via nuclear factor I A. Knockout of IGF1 in CAFs or nuclear factor erythroid 2-related factor 2 in ESCC cells suppressed the tumor growth and chemotherapy resistance induced by CAFs in vivo. Importantly, patients with high proportions of hsCAFs showed poor survival and a worse response to chemotherapy. In summary, these findings identify a hsCAF subpopulation generated by interplay between cancer cells and CAFs under hypoxic conditions that promotes ESCC stemness and reveal targeting hsCAFs as an effective therapeutic strategy against chemotherapy-resistant ESCC. Significance: A hypoxic microenvironment and cancer cells cooperate to induce a senescent fibroblast subset that supports tumor stemness, suggesting that targeting this cancer-associated fibroblast subpopulation is a potential therapeutic strategy to overcome chemoresistance.
Collapse
Affiliation(s)
- Zhengjie Ou
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Liang Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Xinjie Chen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Tianyuan Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Guoyu Cheng
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Rucheng Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Wen Tan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- CAMS Oxford Institute, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
19
|
Cui B, Luo H, He B, Liu X, Lv D, Zhang X, Su K, Zheng S, Lu J, Wang C, Yang Y, Zhao Z, Liu X, Wang X, Zhao Y, Nie X, Jiang Y, Zhang Z, Liu C, Chen X, Cai A, Lv Z, Liu Z, An F, Zhang Y, Yan Q, Kelley KW, Xu G, Xu L, Liu Q, Peng F. Gut dysbiosis conveys psychological stress to activate LRP5/β-catenin pathway promoting cancer stemness. Signal Transduct Target Ther 2025; 10:79. [PMID: 40038255 PMCID: PMC11880501 DOI: 10.1038/s41392-025-02159-1] [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] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 01/02/2025] [Accepted: 01/23/2025] [Indexed: 03/06/2025] Open
Abstract
Psychological stress causes gut microbial dysbiosis and cancer progression, yet how gut microbiota determines psychological stress-induced tumor development remains unclear. Here we showed that psychological stress promotes breast tumor growth and cancer stemness, an outcome that depends on gut microbiota in germ-free and antibiotic-treated mice. Metagenomic and metabolomic analyses revealed that psychological stress markedly alters the composition and abundance of gut microbiota, especially Akkermansia muciniphila (A. muciniphila), and decreases short-chain fatty acid butyrate. Supplement of active A. muciniphila, butyrate or a butyrate-producing high fiber diet dramatically reversed the oncogenic property and anxiety-like behavior of psychological stress in a murine spontaneous tumor model or an orthotopic tumor model. Mechanistically, RNA sequencing analysis screened out that butyrate decreases LRP5 expression to block the activation of Wnt/β-catenin signaling pathway, dampening breast cancer stemness. Moreover, butyrate as a HDAC inhibitor elevated histone H3K9 acetylation level to transcriptionally activate ZFP36, which further accelerates LRP5 mRNA decay by binding adenine uridine-rich (AU-rich) elements of LRP5 transcript. Clinically, fecal A. muciniphila and serum butyrate were inversely correlated with tumoral LRP5/β-catenin expression, poor prognosis and negative mood in breast cancer patients. Altogether, our findings uncover a microbiota-dependent mechanism of psychological stress-triggered cancer stemness, and provide both clinical biomarkers and potential therapeutic avenues for cancer patients undergoing psychological stress.
Collapse
Affiliation(s)
- Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Huandong Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Bin He
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xinyu Liu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, China
| | - Dekang Lv
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Keyu Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Sijia Zheng
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, China
| | - Jinxin Lu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yuqing Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhuoran Zhao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xianxian Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xu Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yingrui Zhao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiaoshan Nie
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yuanyuan Jiang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Ziyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Congcong Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xinyi Chen
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Anqi Cai
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhumeng Lv
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhihang Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Fan An
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yunkun Zhang
- Department of Pathology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Qiulong Yan
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Keith W Kelley
- Department of Pathology, College of Medicine and Department of Animal Sciences, College of ACES, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Guowang Xu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, China
| | - Lingzhi Xu
- Department of Oncology, the Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.
| | - Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| |
Collapse
|
20
|
Wei X, He Y, Yu Y, Tang S, Liu R, Guo J, Jiang Q, Zhi X, Wang X, Meng D. The Multifaceted Roles of BACH1 in Disease: Implications for Biological Functions and Therapeutic Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412850. [PMID: 39887888 PMCID: PMC11905017 DOI: 10.1002/advs.202412850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/22/2024] [Indexed: 02/01/2025]
Abstract
BTB domain and CNC homolog 1 (BACH1) belongs to the family of basic leucine zipper proteins and is expressed in most mammalian tissues. It can regulate its own expression and play a role in transcriptionally activating or inhibiting downstream target genes. It has a crucial role in various biological processes, such as oxidative stress, cell cycle, heme homeostasis, and immune regulation. Recent research highlights BACH1's significant regulatory roles in a series of conditions, including stem cell pluripotency maintenance and differentiation, growth, senescence, and apoptosis. BACH1 is closely associated with cardiovascular diseases and contributes to angiogenesis, atherosclerosis, restenosis, pathological cardiac hypertrophy, myocardial infarction, and ischemia/reperfusion (I/R) injury. BACH1 promotes tumor cell proliferation and metastasis by altering tumor metabolism and the epithelial-mesenchymal transition phenotype. Moreover, BACH1 appears to show an adverse role in diseases such as neurodegenerative diseases, gastrointestinal disorders, leukemia, pulmonary fibrosis, and skin diseases. Inhibiting BACH1 may be beneficial for treating these diseases. This review summarizes the role of BACH1 and its regulatory mechanism in different cell types and diseases, proposing that precise targeted intervention of BACH1 may provide new strategies for human disease prevention and treatment.
Collapse
Affiliation(s)
- Xiangxiang Wei
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Yunquan He
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Yueyang Yu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Sichong Tang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Ruiwen Liu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Jieyu Guo
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Qingjun Jiang
- Department of Vascular & Endovascular SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Xiuling Zhi
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Xinhong Wang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Dan Meng
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| |
Collapse
|
21
|
Wang X, Feng JK, Mao FF, Hou YC, Zhang YQ, Liu LH, Wei Q, Sun JX, Liu C, Shi J, Cheng SQ. Prognostic and Immunotherapeutic Predictive Value of CAD Gene in Hepatocellular Carcinoma: Integrated Bioinformatics and Experimental Analysis. Mol Biotechnol 2025; 67:1240-1255. [PMID: 38683442 DOI: 10.1007/s12033-024-01125-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/27/2024] [Indexed: 05/01/2024]
Abstract
Hepatocellular carcinoma (HCC) is a common type of cancer that ranks first in cancer-associated death worldwide. Carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD) are the key components of the pyrimidine pathway, which promotes cancer development. However, the function of CAD in HCC needs to be clarified. In this study, the clinical and transcriptome data of 424 TCGA-derived HCC cases were analyzed. The results demonstrated that high CAD expression was associated with poor prognosis in HCC patients. The effect of CAD on HCC was then investigated comprehensively using GO annotation analysis, KEGG enrichment analysis, Gene Set Enrichment Analysis (GSEA), and CIBERSORT algorithm. The results showed that CAD expression was correlated with immune checkpoint inhibitors and immune cell infiltration. In addition, low CAD levels in HCC patients predicted increased sensitivity to anti-CTLA4 and PD1, while HCC patients with high CAD expression exhibited high sensitivity to chemotherapeutic and molecular-targeted agents, including gemcitabine, paclitaxel, and sorafenib. Finally, the results from clinical sample suggested that CAD expression increased remarkably in HCC compared with non-cancerous tissues. Loss of function experiments demonstrated that CAD knockdown could significantly inhibit HCC cell growth and migration both in vitro and in vivo. Collectively, the results indicated that CAD is a potential oncogene during HCC metastasis and progression. Therefore, CAD is recommended as a candidate marker and target for HCC prediction and treatment.
Collapse
Affiliation(s)
- Xu Wang
- Cancer Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200433, China
| | - Jin-Kai Feng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200433, China
| | - Fei-Fei Mao
- Tongji University Cancer Center, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yu-Chao Hou
- Cancer Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200433, China
| | - Yu-Qing Zhang
- Cancer Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Li-Heng Liu
- Cancer Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200433, China
| | - Qian Wei
- The First Clinical Medicine School, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ju-Xian Sun
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200433, China
| | - Chang Liu
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200433, China
| | - Jie Shi
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200433, China.
| | - Shu-Qun Cheng
- Cancer Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China.
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200433, China.
- Tongji University Cancer Center, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
22
|
Deng X, Jiao Y, Hao H, Guo Z, An G, Zhang W, Xue D, Han S. Dandelion extract suppresses the stem-like properties of triple-negative breast cancer cells by regulating CUEDC2/β-catenin/OCT4 signaling axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119408. [PMID: 39864604 DOI: 10.1016/j.jep.2025.119408] [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: 12/04/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Triple-negative breast cancer (TNBC) represents the most aggressive subtype of breast cancer, featuring a high proportion of cancer stem cells (CSCs) and the poorest clinical outcomes. Taraxacum mongolicum Hand. -Mazz., widely recognized as dandelion, is a traditional medicinal herb that has demonstrated promising anti-TNBC potential. However, the efficacy of dandelion in anti-TNBC stem-like properties remains to be elucidated. AIM OF THE STUDY The aim was to examine the impact of dandelion extract on the stemness properties of TNBC and to delineate the underlying mechanisms. MATERIALS AND METHODS UHPLC-Q-Orbitrap HRMS was employed to characterize the components present in dandelion extract. Network pharmacology was utilized to explore the impact of dandelion-derived compounds on the molecular pathways associated with TNBC. The assessment of TNBC stem-like properties was conducted through mammosphere formation assays and flow cytometry analysis. Western blotting, qRT-PCR, and immunofluorescence were employed to investigate the mechanisms of dandelion extract. 4T1-luc xenograft tumor model was used to assess the anti-tumor effect of dandelion extract in vivo. IVIS imaging technology was used to monitor lung metastasis. RESULTS In this study, pharmacological network analysis revealed the potential regulatory effects of dandelion extract on TNBC stemness. Dandelion extract disrupts the stem-like properties in MDA-MB-231 and MDA-MB-468 cell lines via reducing ALDH + cells proportion, impeding mammosphere formation, and downregulating CSC-related markers, including SOX2, SOX9, NANOG, and FOXM1. Furthermore, CUE domain containing protein 2 (CUEDC2) promotes the maintenance of TNBC stemness and contributes to the anti-stemness effects of dandelion extract. Mechanistically, dandelion extract inhibits CUEDC2-mediated nuclear translocation of β-catenin, thereby reducing the transcriptional activity of OCT4. In vivo, dandelion extract suppresses tumor growth, lung metastasis, and decreases the expression of CSC-related markers. CONCLUSION These findings suggest that dandelion extract inhibits TNBC stem-like properties via modulating the CUEDC2/β-catenin/OCT4 signaling axis, highlighting its potential as a therapeutic option for TNBC.
Collapse
Affiliation(s)
- Xinxin Deng
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yanna Jiao
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Huifeng Hao
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zhengwang Guo
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Guo An
- Department of Laboratory Animal, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wenlong Zhang
- Department of Laboratory Animal, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Dong Xue
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Shuyan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China; Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
23
|
Liu J, Yao L, Yang Y, Ma J, You R, Yu Z, Du P. A novel stemness-related lncRNA signature predicts prognosis, immune infiltration and drug sensitivity of clear cell renal cell carcinoma. J Transl Med 2025; 23:238. [PMID: 40016772 PMCID: PMC11869577 DOI: 10.1186/s12967-025-06251-6] [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: 07/28/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a prevalent urogenital malignancy characterized by heterogeneous patterns. Stemness is a pivotal factor in tumor progression, recurrence, and metastasis. Nevertheless, the impact of stemness-related long non-coding RNAs (SRlncRNAs) on the prognosis of ccRCC remains elusive. In this study, we aimed to delve into the SRlncRNAs of ccRCC and develop a signature for risk stratification and prognosis prediction. METHOD Gene-expression and clinical data were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. We calculated RNA stemness scores (RNAss) for the samples to evaluate their stemness. SRlncRNAs and stemness-related mRNAs (SRmRNAs) in ccRCC were identified through weighted correlation network analysis (WGCNA), which employed sophisticated statistical methodologies to identify interconnected modules of related genes. Enrichment analysis was performed to explore the potential functions of SRmRNAs. Multiple machine learning algorithms were employed to construct a prognostic signature. Samples from TCGA-KIRC and GSE29609 cohorts were designated as the training and validation cohorts, respectively. Based on their risk scores, samples were stratified into low- and high-risk groups. Prognosis analysis, immune infiltration assessment, drug sensitivity prediction, mutation landscape, and gene set enrichment analysis (GSEA) were conducted to investigate the distinct characteristics of the low- and high-risk groups. Additionally, a web-based calculator was developed to facilitate clinical application. Expression and effects of SRlncRNAs in ccRCC were further corroborated through the utilization of single-cell RNA-seq (scRNA-seq), as well as in vitro and in vivo experiments. RESULTS SRlncRNAs and SRmRNAs were identified based on RNAss and WGCNA. The least absolute shrinkage and selection operator (LASSO) in combination with multivariate Cox regression was selected as the optimal approach. Six SRlncRNAs were used to construct the prognostic signature. Samples in the low- and high-risk groups exhibited distinct characteristics in terms of prognosis, GSEA pathways, immune infiltration profiles, drug sensitivity, and mutation status. A nomogram and a web-based calculator were developed to facilitate the clinical application of the model. ScRNA-seq and RT-qPCR demonstrated the differential expression of SRlncRNAs between ccRCC tumors and normal tissues. In vitro and in vivo experiments demonstrated that downregulation of EMX2OS and LINC00944 affected the proliferation, migration, invasion, apoptosis, and metastasis of ccRCC cells. CONCLUSION We uncovered the crucial associations between SRlncRNAs and the prognosis of ccRCC. By leveraging these findings, we developed a novel SRlncRNA-related signature and a user-friendly web calculator. This signature holds great potential in facilitating risk stratification and guiding tailored treatment strategies for ccRCC patients. Both in vitro and in vivo experiments confirmed the role of SRlncRNAs in the progression of ccRCC.
Collapse
Affiliation(s)
- Jia Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lin Yao
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Yong Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jinchao Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ruijian You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ziyi Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Peng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
24
|
He Q, Xiong Y, Yang X, Yu Y, Chen Z. Molecular subtyping combined with multiomics analysis to study correlation between TACE refractoriness and tumor stemness in hepatocellular carcinoma. Discov Oncol 2025; 16:197. [PMID: 39961903 PMCID: PMC11832877 DOI: 10.1007/s12672-025-01955-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Transarterial chemoembolization (TACE) refractoriness is a significant challenge in treating intermediate to advanced-stage hepatocellular carcinoma (HCC). A few studies suggest that liver cancer stem cells (LCSCs) may be associated with TACE refractoriness. This study aims to explore the potential correlation between TACE refractoriness and HCC stemness, highlighting its clinical significance. METHODS This research encompassed the analysis of diverse HCC datasets, including RNA-sequencing, microarray, single-cell RNA-sequencing, and clinical cohorts. We identified common genes between TACE refractoriness and tumor stemness (TSGs). Unsupervised clustering was employed to classify HCC patients into different clusters based on TSGs (TRS clusters). The study explored the differences in clinical prognosis, biological characteristics, genomic variations, immune landscapes, and treatment responses among the TRS clusters. RESULTS Patients with TACE-refractoriness demonstrated significantly higher tumor stemness. Our study identified 33 TSGs and established two TRS clusters, including C1 and C2. C1 was associated with TACE refractoriness, elevated tumor stemness, and poorer prognosis. Genomic alterations were found to be significantly different between the TRS clusters. The C1 exhibited signs of immunosuppression and lower activity of immune effector cells, while the C2 had a more robust immune response and higher level of immune cell presence. Single-cell RNA-seq revealed distinct cell type characteristics in each subtypes, with the C1 showing a higher proportion of stem cells and malignant cells. CONCLUSION Our findings establish a connection between TACE refractoriness and tumor stemness in HCC, proposing a novel subtype classification to guide personalized treatment. Insights gained may facilitate overcoming TACE refractoriness and the development of innovative therapies.
Collapse
Affiliation(s)
- Qifan He
- Department of Radiology, Haining People's Hospital, No.2 Qianjiang West Road, Haining, 314400, China
| | - Yue Xiong
- Department of Radiology, Haining People's Hospital, No.2 Qianjiang West Road, Haining, 314400, China
| | - Xiaoyu Yang
- Department of Radiology, Haining People's Hospital, No.2 Qianjiang West Road, Haining, 314400, China
| | - Yihui Yu
- Department of Radiology, Haining People's Hospital, No.2 Qianjiang West Road, Haining, 314400, China
| | - Zhonghua Chen
- Department of Radiology, Haining People's Hospital, No.2 Qianjiang West Road, Haining, 314400, China.
| |
Collapse
|
25
|
Lu X, Du W, Zhou J, Li W, Fu Z, Ye Z, Chen G, Huang X, Guo Y, Liao J. Integrated genomic analysis of the stemness index signature of mRNA expression predicts lung adenocarcinoma prognosis and immune landscape. PeerJ 2025; 13:e18945. [PMID: 39959839 PMCID: PMC11830367 DOI: 10.7717/peerj.18945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/16/2025] [Indexed: 02/18/2025] Open
Abstract
mRNA expression-based stemness index (mRNAsi) has been used for prognostic assessment in various cancers, but its application in lung adenocarcinoma (LUAD) is limited, which is the focus of this study. Low mRNAsi in LUAD predicted a better prognosis. Eight genes (GNG7, EIF5A, ANLN, FKBP4, GAPDH, GNPNAT1, E2F7, CISH) associated with mRNAsi were screened to establish a risk model. The differentially expressed genes between the high and low risk groups were mainly enriched in the metabolism, cell cycle functions pathway. The low risk score group had higher immune cell scores. Patients with lower TIDE scores in the low risk group had better immunotherapy outcomes. In addition, risk score was effective in assessing drug sensitivity of LUAD. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) data showed that eight genes were differentially expressed in LUAD cell lines, and knockdown of EIF5A reduced the invasion and migration ability of LUAD cells. This study designed a risk model based on the eight mRNAsi-related genes for predicting LUAD prognosis. The model accurately predicted the prognosis and survival of LUAD patients, facilitating the assessment of the sensitivity of patients to immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Xingzhao Lu
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
- Department of Medical Oncology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Wei Du
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Jianping Zhou
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Weiyang Li
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Zhimin Fu
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Zhibin Ye
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Guobiao Chen
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Xian Huang
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Yuliang Guo
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Jingsheng Liao
- Department of Medical Oncology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| |
Collapse
|
26
|
Verona F, Di Bella S, Schirano R, Manfredi C, Angeloro F, Bozzari G, Todaro M, Giannini G, Stassi G, Veschi V. Cancer stem cells and tumor-associated macrophages as mates in tumor progression: mechanisms of crosstalk and advanced bioinformatic tools to dissect their phenotypes and interaction. Front Immunol 2025; 16:1529847. [PMID: 39981232 PMCID: PMC11839637 DOI: 10.3389/fimmu.2025.1529847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Cancer stem cells (CSCs) are a small subset within the tumor mass significantly contributing to cancer progression through dysregulation of various oncogenic pathways, driving tumor growth, chemoresistance and metastasis formation. The aggressive behavior of CSCs is guided by several intracellular signaling pathways such as WNT, NF-kappa-B, NOTCH, Hedgehog, JAK-STAT, PI3K/AKT1/MTOR, TGF/SMAD, PPAR and MAPK kinases, as well as extracellular vesicles such as exosomes, and extracellular signaling molecules such as cytokines, chemokines, pro-angiogenetic and growth factors, which finely regulate CSC phenotype. In this scenario, tumor microenvironment (TME) is a key player in the establishment of a permissive tumor niche, where CSCs engage in intricate communications with diverse immune cells. The "oncogenic" immune cells are mainly represented by B and T lymphocytes, NK cells, and dendritic cells. Among immune cells, macrophages exhibit a more plastic and adaptable phenotype due to their different subpopulations, which are characterized by both immunosuppressive and inflammatory phenotypes. Specifically, tumor-associated macrophages (TAMs) create an immunosuppressive milieu through the production of a plethora of paracrine factors (IL-6, IL-12, TNF-alpha, TGF-beta, CCL1, CCL18) promoting the acquisition by CSCs of a stem-like, invasive and metastatic phenotype. TAMs have demonstrated the ability to communicate with CSCs via direct ligand/receptor (such as CD90/CD11b, LSECtin/BTN3A3, EPHA4/Ephrin) interaction. On the other hand, CSCs exhibited their capacity to influence immune cells, creating a favorable microenvironment for cancer progression. Interestingly, the bidirectional influence of CSCs and TME leads to an epigenetic reprogramming which sustains malignant transformation. Nowadays, the integration of biological and computational data obtained by cutting-edge technologies (single-cell RNA sequencing, spatial transcriptomics, trajectory analysis) has significantly improved the comprehension of the biunivocal multicellular dialogue, providing a comprehensive view of the heterogeneity and dynamics of CSCs, and uncovering alternative mechanisms of immune evasion and therapeutic resistance. Moreover, the combination of biology and computational data will lead to the development of innovative target therapies dampening CSC-TME interaction. Here, we aim to elucidate the most recent insights on CSCs biology and their complex interactions with TME immune cells, specifically TAMs, tracing an exhaustive scenario from the primary tumor to metastasis formation.
Collapse
Affiliation(s)
- Francesco Verona
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Sebastiano Di Bella
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Roberto Schirano
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Camilla Manfredi
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Francesca Angeloro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Giulia Bozzari
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
- Azienda Ospedaliera Universitaria Policlinico “Paolo Giaccone” (AOUP), Palermo, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
- Istituto Pasteur, Fondazione Cenci-Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Giorgio Stassi
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Veronica Veschi
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| |
Collapse
|
27
|
Li S, Gao S, Qin L, Ding C, Qu J, Cui Y, Qiang L, Yin S, Zheng X, Meng H. Micropapillary structure: A natural tumor collective invasion model with enhanced stem-like properties. Cancer Sci 2025; 116:308-321. [PMID: 39568148 PMCID: PMC11786311 DOI: 10.1111/cas.16396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
Cancer stem cells aggregate to form clusters, which have enhanced stem-like properties and metastasis potential. However, the molecular mechanisms underlying the formation of cancer stem cell cluster-like structures with acquisition of stronger invasion and metastasis abilities remain unclear. Micropapillary carcinoma (MPC) is a subpopulation of small, merulioid, inverted, nonfibrous vascular clusters floating in the stroma present in a range of solid malignant tumors and characterized by frequent vascular/lymphatic vessel invasion and lymph node metastasis. Our results showed that these cell clusters exhibit a stem cell phenotype, supporting the premise that MPC may serve as a promising solid tumor model for studying invasion and metastasis of cancer stem cell clusters. In this review, we discuss the latest advances in MPC research and targeted therapy, focusing on analysis of their stem-like characteristics, mapping their multiomics characteristics, and elucidating the vascular and immune microenvironment of MPC. The existing MPC organoid model was employed to explore potential breakthroughs in targeted therapy and immunotherapy for cancer stem cell clusters.
Collapse
Affiliation(s)
- Sisi Li
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Shuangshu Gao
- Department of PathologyHarbin Medical UniversityHarbinChina
| | - Ling Qin
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Caixia Ding
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Jinghui Qu
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Yifei Cui
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Lixia Qiang
- Department of Respiratory MedicineThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Shengjie Yin
- Department of Medical OncologyMunicipal Hospital of ChifengChifengChina
| | - Xiaoyu Zheng
- Department of AnesthesiologyHarbin Medical University Cancer HospitalHarbinChina
| | - Hongxue Meng
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| |
Collapse
|
28
|
Huang X, Chen W, Wang Y, Shytikov D, Wang Y, Zhu W, Chen R, He Y, Yang Y, Guo W. Canonical and noncanonical NOTCH signaling in the nongenetic resistance of cancer: distinct and concerted control. Front Med 2025; 19:23-52. [PMID: 39745621 DOI: 10.1007/s11684-024-1107-1] [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: 04/19/2024] [Accepted: 09/18/2024] [Indexed: 02/27/2025]
Abstract
Therapeutic resistance in cancer is responsible for numerous cancer deaths in clinical practice. While target mutations are well recognized as the basis of genetic resistance to targeted therapy, nontarget mutation resistance (or nongenetic resistance) remains poorly characterized. Despite its complex and unintegrated mechanisms in the literature, nongenetic resistance is considered from our perspective to be a collective response of innate or acquired resistant subpopulations in heterogeneous tumors to therapy. These subpopulations, e.g., cancer stem-like cells, cancer cells with epithelial-to-mesenchymal transition, and drug-tolerant persisters, are protected by their resistance traits at cellular and molecular levels. This review summarizes recent advances in the research on resistant populations and their resistance traits. NOTCH signaling, as a central regulator of nongenetic resistance, is discussed with a special focus on its canonical maintenance of resistant cancer cells and noncanonical regulation of their resistance traits. This novel view of canonical and noncanonical NOTCH signaling pathways is translated into our proposal of reshaping therapeutic strategies targeting NOTCH signaling in resistant cancer cells. We hope that this review will lead researchers to study the canonical and noncanonical arms of NOTCH signaling as an integrated resistant mechanism, thus promoting the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xianzhe Huang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Wenwei Chen
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Yanyan Wang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Dmytro Shytikov
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Yanwen Wang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Wangyi Zhu
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Ruyi Chen
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Yuwei He
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Yanjia Yang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Wei Guo
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China.
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Biomedical and Health Translational Research Center of Zhejiang Province, Jiaxing, 314400, China.
| |
Collapse
|
29
|
Zhou F, Guo H, Xia Y, Le X, Tan DSW, Ramalingam SS, Zhou C. The changing treatment landscape of EGFR-mutant non-small-cell lung cancer. Nat Rev Clin Oncol 2025; 22:95-116. [PMID: 39614090 DOI: 10.1038/s41571-024-00971-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/01/2024]
Abstract
The discovery of the association between EGFR mutations and the efficacy of EGFR tyrosine-kinase inhibitors (TKIs) has revolutionized the treatment paradigm for patients with non-small-cell lung cancer (NSCLC). Currently, third-generation EGFR TKIs, which are often characterized by potent central nervous system penetrance, are the standard-of-care first-line treatment for advanced-stage EGFR-mutant NSCLC. Rational combinations of third-generation EGFR TKIs with anti-angiogenic drugs, chemotherapy, the EGFR-MET bispecific antibody amivantamab or local tumour ablation are being investigated as strategies to delay drug resistance and increase clinical benefit. Furthermore, EGFR TKIs are being evaluated in patients with early stage or locally advanced EGFR-mutant NSCLC, with the ambitious aim of achieving cancer cure. Despite the inevitable challenge of acquired resistance, emerging treatments such as new TKIs, antibody-drug conjugates, new immunotherapeutic approaches and targeted protein degraders have shown considerable promise in patients with progression of EGFR-mutant NSCLC on or after treatment with EGFR TKIs. In this Review, we describe the current first-line treatment options for EGFR-mutant NSCLC, provide an overview of the mechanisms of acquired resistance to third-generation EGFR TKIs and explore novel promising treatment strategies. We also highlight potential avenues for future research that are aimed at improving the survival outcomes of patients with this disease.
Collapse
Affiliation(s)
- Fei Zhou
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haoyue Guo
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yang Xia
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuning Le
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Duke-NUS Medical School, Singapore, Singapore
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA, USA
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
30
|
T C D, N K N, Pushparaj C, Narayanasamy A, Manickam P, Thiruvenkataswamy S, Sennimalai R. Novel therapeutic approaches targeting cancer stem cells: Unveiling new frontiers in breast cancer treatment. Pathol Res Pract 2025; 266:155800. [PMID: 39808859 DOI: 10.1016/j.prp.2024.155800] [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: 10/17/2024] [Revised: 12/13/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025]
Abstract
Breast cancer remains the leading cause of mortality among women with cancer. This article delves into the intricate relationship between breast cancer and cancer stem cells (CSCs), emphasizing advanced methods for their identification and isolation. The key isolation techniques, such as the mammosphere formation assay, surface marker identification, Side Population assay, and Aldehyde Dehydrogenase assay, are critically examined. Furthermore, the review analyzes CSC-specific molecular signaling pathways, focusing on actionable targets like CD44/CD24, Nanog, and Oct4. The potential of targeted therapies and small molecules that disrupt these pathways is explored. Additionally, the review highlights immunotherapy strategies against CSCs, focusing on resistance mechanisms and the critical role of precision medicine. The study investigates how precision medicine enhances therapeutic outcomes by targeting specific CSC biomarkers. This comprehensive analysis offers insights into recent advancements and emerging strategies in breast cancer treatment, pointing toward future therapeutic innovations.
Collapse
Affiliation(s)
- Deeptha T C
- Department of Zoology, PSGR Krishnammal College for Women, Coimbatore, India
| | - Nabeela N K
- Department of Zoology, PSGR Krishnammal College for Women, Coimbatore, India
| | | | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, India
| | - Paulpandi Manickam
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, India
| | | | - Ramya Sennimalai
- Department of Zoology (PG), Vellalar College for Women, Erode, India.
| |
Collapse
|
31
|
Liu L, Han F, Deng M, Han Q, Lai M, Zhang H. Crosstalk between GLTSCR1-deficient endothelial cells and tumour cells promotes colorectal cancer development by activating the Notch pathway. Cell Death Differ 2025:10.1038/s41418-025-01450-6. [PMID: 39870803 DOI: 10.1038/s41418-025-01450-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 12/17/2024] [Accepted: 01/21/2025] [Indexed: 01/29/2025] Open
Abstract
Cancer stem cells (CSCs) typically reside in perivascular niches, but whether endothelial cells of blood vessels influence the stemness of cancer cells remains poorly understood. This study revealed that endothelial cell-specific GLTSCR1 deletion promotes colorectal cancer (CRC) tumorigenesis and metastasis by increasing cancer cell stemness. Mechanistically, knocking down GLTSCR1 induces the transformation of endothelial cells into tip cells by regulating the expression of Neuropilin-1 (NRP1), thereby increasing the direct contact and interaction between endothelial cells and tumour cells. In addition, GLTSCR1 inhibits JAG1 transcription by competing with acetylated p65(Lys-310) to bind to the BRD4 interaction site. Therefore, GLTSCR1 deficiency increases JAG1 expression in endothelial cells. Subsequently, increased JAG1 levels on tip cell membranes bind to Notch on CRC cell membranes, activating the Notch signalling pathway in tumour cells and increasing CRC cell stemness. Taken together, our findings highlight the roles of endothelial cells in CRC development.
Collapse
Affiliation(s)
- Lu Liu
- Department of Pathology and International Institutes of Medicine, The Fourth Affiliated Hospital (Yiwu), Zhejiang University School of Medicine, Hangzhou, 310058, China
- Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, 310058, China
| | - Fengyan Han
- School of Basic Medical Sciences, The Fourth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Mengli Deng
- Department of Pathology and International Institutes of Medicine, The Fourth Affiliated Hospital (Yiwu), Zhejiang University School of Medicine, Hangzhou, 310058, China
- Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, 310058, China
| | - Qizheng Han
- Department of Pathology and International Institutes of Medicine, The Fourth Affiliated Hospital (Yiwu), Zhejiang University School of Medicine, Hangzhou, 310058, China
- Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, 310058, China
| | - Maode Lai
- Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, 310058, China.
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Honghe Zhang
- Department of Pathology and International Institutes of Medicine, The Fourth Affiliated Hospital (Yiwu), Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, 310058, China.
| |
Collapse
|
32
|
Zhang W, Xiang Y, Guo Q, Wang X, Zhang L, Guo J, Cong R, Yu W, Liang XJ, Zhang J, Liu D. Multi-phoretic nanomotor with consistent motion direction for enhanced cancer therapy. Acta Biomater 2025; 191:352-368. [PMID: 39586348 DOI: 10.1016/j.actbio.2024.11.037] [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: 07/17/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Nanomotors have emerged as promising candidates for the deep penetration of loaded drugs into cancer stem cells (CSCs) located within the core of tumor tissues. A crucial factor in maximizing the clinical potential of nanomotors lies in their ability to respond dynamically to various stimuli in the tumor microenvironment. By adjusting their propulsion mechanisms in response to various stimuli, nanomotors can maintain directional movement, thus improving drug distribution and therapeutic efficacy. In this study, we present the design of a pH-responsive multi-phoretic propelled Janus nanomotor, comprising a SiO2@Pt core@shell nanosphere and half-wrapped acrylic acid polymers (PAA)-conjugated gold (Au) nanoparticles (JMSNs@Pt@P-Au). The JMSNs@Pt@P-Au catalyze endogenous H2O2 into O2, propelling the nanomotors into solid tumors. Within the tumor microenvironment, the contraction of PAA triggers contact between the Au and Pt layers, facilitating self-electrophoresis propulsion. Simultaneously, a local thermal gradient is generated on the Au layer under near-infrared light irradiation, propelling the nanomotor through thermophoresis. Exploiting the unique structure of JMSNs@Pt@P-Au, the driving forces generated by H2O2 catalysis, self-electrophoresis, and thermophoresis exhibit consistent motion directions. This consistency not only provides thrust for deep penetration but also enhances their targeted therapeutic efficiency against CSCs in vivo. This combination of nanomotor-driven power sources holds significant potential for designing intelligent, active drug delivery systems for effective CSC-targeted cancer therapy. STATEMENT OF SIGNIFICANCE: Deep penetration of nanomedicine in solid tumor tissue and cells is still an important challenge that restricts the therapeutic effect. Multiple-propelled nanomotors have been confirmed to be self-propulsive that overcome the limited penetration in solid tumor. However, their effective translation toward clinical applications is limited due to the inability to alter their propelled mechanisms in response to the actual physiological environment, resulting in speed and inconsistent movement directions. In this work, we designed a multi-phoretic propelled Janus nanomotor (JMSNs@Pt@P-Au) that exhibited three propelled mechanisms in response to the changes of pH value. Noteworthy is their heightened speed and remarkable tumor tissue penetration observed in vitro and in vivo without adverse effects. Such multi-phoretic propulsion offers considerable promise for developing advanced nanomachines with a stimuli-responsive switch of propulsion modes in biomedical applications.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province & College of Chemistry and Materials Science, Hebei University, Baoding, 071002, PR China
| | - Yangyang Xiang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province & College of Chemistry and Materials Science, Hebei University, Baoding, 071002, PR China
| | - Qi Guo
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province & College of Chemistry and Materials Science, Hebei University, Baoding, 071002, PR China
| | - Xiaotong Wang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province & College of Chemistry and Materials Science, Hebei University, Baoding, 071002, PR China
| | - Lukai Zhang
- College of Physics Science & Technology, Hebei University, Baoding, 071002, PR China
| | - Jiaxin Guo
- College of Physics Science & Technology, Hebei University, Baoding, 071002, PR China
| | - Ridong Cong
- College of Physics Science & Technology, Hebei University, Baoding, 071002, PR China
| | - Wei Yu
- College of Physics Science & Technology, Hebei University, Baoding, 071002, PR China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, PR China
| | - Jinchao Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province & College of Chemistry and Materials Science, Hebei University, Baoding, 071002, PR China.
| | - Dandan Liu
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province & College of Chemistry and Materials Science, Hebei University, Baoding, 071002, PR China.
| |
Collapse
|
33
|
Ma D, Liang R, Luo Q, Song G. Pressure loading regulates the stemness of liver cancer stem cells via YAP/BMF signaling axis. J Cell Physiol 2025; 240:e31451. [PMID: 39358905 DOI: 10.1002/jcp.31451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
Cancer stem cells (CSCs) are considered the major cause of the occurrence, progression, chemoresistance/radioresistance, recurrence, and metastasis of cancer. Increased interstitial fluid pressure (IFP) is a key feature of solid tumors. Our previous study showed that the distribution of liver cancer stem cells (LCSCs) correlated with the mechanical heterogeneity within liver cancer tissues. However, the regulation of liver cancer's mechanical microenvironment on the LCSC stemness is not fully understood. Here, we employed a cellular pressure-loading device to investigate the effects of normal IFP (5 mmHg), as well as increased IFP (40 and 200 mmHg) on the stemness of LCSCs. Compared to the control LCSCs (exposure to 5 mmHg pressure loading), the LCSCs exposed to 40 mmHg pressure loading exhibited significantly upregulated expression of CSC markers (CD44, EpCAM, Nanog), enhanced sphere and colony formation capacities, and tumorigenic potential, whereas continuously increased pressure to 200 mmHg suppressed the LCSC characteristics. Mechanistically, pressure loading regulated Yes-associated protein (YAP) activity and Bcl-2 modifying factor (BMF) expression. YAP transcriptionally regulated BMF expression to affect the stemness of LCSCs. Knockdown of YAP and overexpression of BMF attenuated pressure-mediated stemness and tumorgenicity, while YAP-deficient and BMF-deletion recused pressure-dependent stemness on LCSCs, suggesting the involvement of YAP/BMF signaling axis in this process. Together, our findings provide a potential target for overcoming the stemness of CSCs and elucidate the significance of increased IFP in cancer progression.
Collapse
Affiliation(s)
- Di Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Rui Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
34
|
Yi K, Chen W, Zhou X, Xie C, Zhong C, Zhu J. Bisphenol S exposure promotes stemness of triple-negative breast cancer cells via regulating Gli1-mediated Sonic hedgehog pathway. ENVIRONMENTAL RESEARCH 2025; 264:120293. [PMID: 39505130 DOI: 10.1016/j.envres.2024.120293] [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: 06/20/2024] [Revised: 10/09/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
Bisphenol S (BPS), one of the most common alternatives for bisphenol A (BPA), has been implied to increase the risk of breast cancer. Triple-negative breast cancer (TNBC) is a highly aggressive type of breast cancer with a poor prognosis. However, the association between BPS and TNBC remains unclear. Cancer stem cells (CSCs) have a crucial role in breast cancer initiation, metastasis, and recurrence. Here, we proposed that BPS, equivalent to the human internal exposure and the environmental concentrations, enhanced CSC-like properties by upregulating sphere formation, self-renewal, the percentage of CD44+/CD24- cells, and the expression of CSC markers. Moreover, BPS promoted the migration, invasion, and epithelial-mesenchymal transition (EMT) in TNBC cells. Mechanistically, BPS activated the Sonic Hedgehog (SHH) signaling pathway in TNBC cells. Molecular docking analysis further showed that BPS upregulated SHH signaling pathway via directly binding Gli1 protein. Furthermore, inhibitor of SHH pathway or Gli1 siRNA attenuated the promoting effects of BPS on stemness, invasion, and migration of TNBC cells. In summary, our data firstly provide evidence that environmentally relevant BPS concentration treatment significantly enhanced TNBC malignant phenotype by activating the Sonic Hedgehog/Gli1 signaling pathway, raising high concerns about the potential population biology hazards of BPS.
Collapse
Affiliation(s)
- Kefan Yi
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiyi Chen
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Zhou
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jianyun Zhu
- Department of Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China.
| |
Collapse
|
35
|
Zhang X, Xie J, Yang Z, Yu CKW, Hu Y, Qin J. Tumour heterogeneity and personalized treatment screening based on single-cell transcriptomics. Comput Struct Biotechnol J 2024; 27:307-320. [PMID: 39877290 PMCID: PMC11773088 DOI: 10.1016/j.csbj.2024.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
According to global cancer statistics for the year 2022, based on updated estimates from the International Agency for Research on Cancer, there were approximately 20 million new cases of cancer in 2022 alongside 9.7 million related deaths. Lung, breast, colorectal, gastric, and liver cancers are the most common types of cancer. Despite advancements in anticancer drugs and optimised chemotherapy regimens that have improved cure rates for malignant tumours, the presence of tumour heterogeneity has resulted in substantial variations among patients in terms of disease progression, clinical response, sensitivity to therapy, and prognosis, posing significant challenges in attaining optimal therapeutic outcomes for each patient. Here, we collected five single-cell transcriptome datasets from patients with lung, breast, colorectal, gastric, and liver cancers and constructed multiple cancer blueprints of tumour cell heterogeneity. By integrating multiple bioinformatics analyses, we explored the biological differences underlying tumour cell heterogeneity at the single-cell level and identified tumour cell subcluster-specific biomarkers and potential therapeutic drugs for each subcluster. Interestingly, although tumour cell subpopulations exhibit dramatic differences within the same cancer type and between different cancers at both the genomic and transcriptomic levels, some demonstrate similar oncogenic pathway activities and phenotypes. Tumour cell subpopulations from the five cancers listed above were classified into three major groups corresponding to different treatment strategies. The findings of this study not only focus on the differences but also on the similarities among tumour cell subpopulations across different cancers, providing new insights for individualised therapy.
Collapse
Affiliation(s)
- Xinying Zhang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Jiajie Xie
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zixin Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Carisa Kwok Wai Yu
- Department of Mathematics, Statistics and Insurance, The Hang Seng University of Hong Kong, Shatin, Hong Kong
| | - Yaohua Hu
- School of Mathematical Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Jing Qin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| |
Collapse
|
36
|
Sun R, Liu K, Pan S, Ye Y, Li N, Chen S, Cui X, Zhang Y, Chen L, Pan J, Hu Z, Luo C, Fan J, Zhou Z, Zhou S, Zhou J. LRP4 mutations promote tumor progression and resistance to anti-PD-1 therapy in recurrent hepatocellular carcinoma. Hepatology 2024:01515467-990000000-01125. [PMID: 39723987 DOI: 10.1097/hep.0000000000001212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND AND AIMS HCC recurrence is a major factor limiting long-term survival and the cause of most deaths in patients with HCC. However, molecular characterization and potential therapeutic targets of recurrent HCC remain mostly unknown. APPROACH AND RESULTS We performed whole-exome sequencing in 63 matched primary and recurrent HCC tumors and combined the data with whole-genome sequencing results in 43 paired samples from our previous study. Sanger sequencing was used to identify all low-density lipoprotein receptor-related protein 4 ( LRP4 ) coding exons in 203 additional patients with recurrent HCC. We identified LRP4 somatic mutations in 7.8% (24/309) of recurrent tumors and only 0.97% (3/309) of primary tumors ( p <0.001). Prognosis after the second liver resection was poorer in patients with an LRP4 mutation. Biofunctional investigations demonstrated that inactivating LRP4 mutations promoted tumor progression and immunosuppression. Mechanistically, mutated LRP4 reduced intratumoral conventional type 1 dendritic cell and CD8 + T cell infiltration by repressing C-C motif chemokine ligand 4 expression and secretion through activation of β-catenin signaling, resulting in resistance to anti-programmed cell death protein-1 therapy. Patients with recurrent HCC carrying an LRP4 mutation did not benefit from anti-programmed cell death protein-1 treatment after their second resection surgery. A β-catenin inhibitor-reversed LRP4-induced resistance to anti-programmed cell death protein-1 therapy in humanized tumor-bearing mice. CONCLUSIONS Our results identified novel LRP4 mutations important in recurrent HCC. Inactivating LRP4 mutations were associated with resistance to anti-programmed cell death protein-1 therapy and could be useful biomarkers for precision therapy in patients with recurrent HCC.
Collapse
Affiliation(s)
- Rongqi Sun
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Kaixuan Liu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Siyuan Pan
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Yuhang Ye
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Ning Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Shuangyi Chen
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Xinyi Cui
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Yuxi Zhang
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Long Chen
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Jingyue Pan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Zhiqiang Hu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Chubin Luo
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Zhengjun Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Shaolai Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
- Greater Bay Area Institute of Precision Medicine, Fudan University, Guangzhou, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Greater Bay Area Institute of Precision Medicine, Fudan University, Guangzhou, China
| |
Collapse
|
37
|
Li X, Han H, Yang K, Li S, Ma L, Yang Z, Zhao YX. Crosstalk between thyroid CSCs and immune cells: basic principles and clinical implications. Front Immunol 2024; 15:1476427. [PMID: 39776907 PMCID: PMC11703838 DOI: 10.3389/fimmu.2024.1476427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Thyroid cancer has become the most common endocrine malignancy. Although the majority of differentiated thyroid cancers have a favorable prognosis, advanced thyroid cancers, iodine-refractory thyroid cancers, and highly malignant undifferentiated carcinomas still face a serious challenge of poor prognosis and even death. Cancer stem cells are recognized as one of the central drivers of tumor evolution, recurrence and treatment resistance. A fresh viewpoint on the oncological aspects of thyroid cancer, including proliferation, invasion, recurrence, metastasis, and therapeutic resistance, has been made possible by the recent thorough understanding of the defining and developing features as well as the plasticity of thyroid cancer stem cells (TCSCs). The above characteristics of TCSCs are complicated and regulated by cell-intrinsic mechanisms (including activation of key stem signaling pathways, somatic cell dedifferentiation, etc.) and cell-extrinsic mechanisms. The complex communication between TCSCs and the infiltrating immune cell populations in the tumor microenvironment (TME) is a paradigm for cell-extrinsic regulators. This review introduces the current advances in the studies of TCSCs, including the origin of TCSCs, the intrinsic signaling pathways regulating the stemness of TCSCs, and emerging biomarkers; We further highlight the underlying principles of bidirectional crosstalk between TCSCs and immune cell populations driving thyroid cancer progression, recurrence, or metastasis, including the specific mechanisms by which immune cells maintain the stemness and other properties of TCSCs and how TCSCs reshape the immune microenvironmental landscape to create an immune evasive and pro-tumorigenic ecological niche. Finally, we outline promising strategies and challenges for targeting key programs in the TCSCs-immune cell crosstalk process to treat thyroid cancer.
Collapse
Affiliation(s)
- Xiaoxiao Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Hengtong Han
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Kaili Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Shouhua Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Libin Ma
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ze Yang
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yong-xun Zhao
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
38
|
Wei Y, Geng S, Chen Q, Chen J, Fan Z, Li Y, Si Y, Yang Y, Liang Z, Jiang J. Protocol for sorting cancer stem cells using a combination of anti-CD133 antibody and lectin cyanovirin-N. STAR Protoc 2024; 5:103443. [PMID: 39565690 PMCID: PMC11617445 DOI: 10.1016/j.xpro.2024.103443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/27/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024] Open
Abstract
CD133 is widely used as a marker to isolate cancer stem cells (CSCs). However, the structural ambiguity of N-glycan of CD133 limits its application in the isolation of CSCs. Here, we present a protocol to sort CSCs from tumor samples by combining CD133 with α-1,2-high-mannose type glycan chains. We describe steps for purifying and biotinylating cyanovirin-N (CVN) proteins and sorting tumor cells and CD133+α-1,2-Man+ (α-1,2-linked mannose+) cells to provide more specific markers for CSCs. For complete details on the use and execution of this protocol, please refer to Wei et al.1.
Collapse
Affiliation(s)
- Yuanyan Wei
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China.
| | - Shuting Geng
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Qihang Chen
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Jiayue Chen
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Zhijun Fan
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Yi Li
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Yu Si
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Yuerong Yang
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Ziwei Liang
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China; Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jianhai Jiang
- NHC Key Laboratory of Glycoconjuates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China.
| |
Collapse
|
39
|
Stajer M, Horacek JM, Kupsa T, Zak P. The role of chemokines and interleukins in acute lymphoblastic leukemia: a systematic review. J Appl Biomed 2024; 22:165-184. [PMID: 40033805 DOI: 10.32725/jab.2024.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/15/2024] [Indexed: 03/05/2025] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood hematological malignancy, but it also affects adult patients with worse prognosis and outcomes. Leukemic cells benefit from protective mechanisms, which are mediated by intercellular signaling molecules - cytokines. Through these signals, cytokines modulate the biology of leukemic cells and their surroundings, enhancing the proliferation, survival, and chemoresistance of the disease. This ultimately leads to disease progression, refractoriness, and relapse, decreasing the chances of curability and overall survival of the patients. Targeting and modulating these pathological processes without affecting the healthy physiology is desirable, offering more possibilities for the treatment of ALL patients, which still remains unsatisfactory in certain cases. In this review, we comprehensively analyze the existing literature and ongoing trials regarding the role of chemokines and interleukins in the biology of ALL. Focusing on the functional pathways, genetic background, and critical checkpoints, we constructed a summary of molecules that are promising for prognostic stratification and mainly therapeutic use. Targeted therapy, including chemokine and interleukin pathways, is a new and promising approach to the treatment of cancer. With the expansion of our knowledge, we are able to uncover a spectrum of new potential checkpoints in order to modulate the disease biology. Several cytokine-related targets are advancing toward clinical application, offering the hope of higher disease response rates to treatment.
Collapse
Affiliation(s)
- Martin Stajer
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Jan M Horacek
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Tomas Kupsa
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Pavel Zak
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| |
Collapse
|
40
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [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: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
41
|
Bae K, Kim DE, Kim JH, Lee JY, Yoon KA. Oncogenic fusion of CD63-BCAR4 contributes cancer stem cell-like properties via ALDH1 activity. Mol Carcinog 2024; 63:2282-2290. [PMID: 39136580 DOI: 10.1002/mc.23808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/13/2024] [Accepted: 07/30/2024] [Indexed: 11/16/2024]
Abstract
Gene fusions are common somatic alterations in cancers, and fusions with tumorigenic features have been identified as novel drivers of cancer and therapeutic targets. Few studies have determined whether the oncogenic ability of fusion genes is related to the induction of stemness in cells. Cancer stem cells (CSCs) are a subset of cells that contribute to cancer progression, metastasis, and recurrence, and are critical components of the aggressive features of cancer. Here, we investigated the CSC-like properties induced by CD63-BCAR4 fusion gene, previously reported as an oncogenic fusion, and its potential contribution for the enhanced metastasis as a notable characteristic of CD63-BCAR4. CD63-BCAR4 overexpression facilitates sphere formation in immortalized bronchial epithelial cells. The significantly enhanced sphere-forming activity observed in tumor-derived cells from xenografted mice of CD63-BCAR4 overexpressing cells was suppressed by silencing of BCAR4. RNA microarray analysis revealed that ALDH1A1 was upregulated in the BCAR4 fusion-overexpressing cells. Increased activity and expression of ALDH1A1 were observed in the spheres of CD63-BCAR4 overexpressing cells compared with those of the empty vector. CD133 and CD44 levels were also elevated in BCAR4 fusion-overexpressing cells. Increased NANOG, SOX2, and OCT-3/4 protein levels were observed in metastatic tumor cells derived from mice injected with CD63-BCAR4 overexpressing cells. Moreover, DEAB, an ALDH1A1 inhibitor, reduced the migration activity induced by CD63-BCAR4 as well as the sphere-forming activity. Our findings suggest that CD63-BCAR4 fusion induces CSC-like properties by upregulating ALDH1A1, which contributes to its metastatic features.
Collapse
Affiliation(s)
- Kieun Bae
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Dong Eon Kim
- College of Health Science, Cheongju University, Cheongju, Republic of Korea
| | - Jin Hee Kim
- College of Health Science, Cheongju University, Cheongju, Republic of Korea
| | - Ja Young Lee
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Kyong-Ah Yoon
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
42
|
Wang Y, Wang Q, Tao S, Li H, Zhang X, Xia Y, Wang Y, Yang C, Sui C. Identification of SPP1 + macrophages in promoting cancer stemness via vitronectin and CCL15 signals crosstalk in liver cancer. Cancer Lett 2024; 604:217199. [PMID: 39216547 DOI: 10.1016/j.canlet.2024.217199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Macrophages play a multifaceted role in cancer biology, with both pro-tumorigenic and anti-tumorigenic functions. Understanding the mechanisms underlying macrophage involvement in cancer progression is essential for the development of therapeutic strategies. Our study analyzed single-cell RNA sequencing data from 12 patients with liver cancer and identified a subpopulation of macrophages characterized by elevated expression of SPP1, which correlates with poor prognosis in liver cancer patients. These SPP1+ macrophages induce upregulation of tumor stemness through a vitronectin (VTN)-dependent paracrine mechanism. Mechanistically, VTN derived from SPP1+ macrophages promote integrin αvβ5/adenosine 5'-monophosphate-activated protein kinase (AMPK)/Yes-associated protein 1 (YAP1)/SYR-box transcription factor 4 (SOX4) signaling, mediating liver tumor stemness and progression. Conversely, CCL15 produced by liver cancer cells drives polarization of M0 macrophages toward an SPP1+ macrophage phenotype, establishing a positive feedback loop of macrophage-tumor stemness. Furthermore, the presence of SPP1+ macrophages confers chemoresistance in liver cancer, and inhibition of the macrophage-tumor feedback loop through targeting integrin αvβ5/YAP1 signaling sensitizes liver cancer cells to chemotherapy. Our study highlights the crucial role of SPP1+ macrophages in liver cancer progression, providing novel insights for clinical liver cancer therapy.
Collapse
Affiliation(s)
- Yizhou Wang
- Department of Hepatic Surgery IV, The Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, PR China; Eastern Hepatobiliary Clinical Research Institute, Third Affiliated Hospital of Navy Medical University, Shanghai, 200438, PR China.
| | - Qing Wang
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Shuangfen Tao
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Haoyu Li
- Department of Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, PR China.
| | - Xiaofeng Zhang
- Department of Hepatic Surgery IV, The Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, PR China; Eastern Hepatobiliary Clinical Research Institute, Third Affiliated Hospital of Navy Medical University, Shanghai, 200438, PR China.
| | - Yong Xia
- Department of Hepatic Surgery IV, The Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, PR China; Eastern Hepatobiliary Clinical Research Institute, Third Affiliated Hospital of Navy Medical University, Shanghai, 200438, PR China.
| | - Yue Wang
- Department of Stem Cell and Regeneration Medicine, Translational Medicine Research Center, Naval Medical University, Shanghai, 200433, PR China; Department of Histology and Embryology, Basic Medicine Collage, Naval Medical University, Shanghai, 200433, PR China; Shanghai Key Laboratory of Cell Engineering, Shanghai, 200062, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200092, PR China.
| | - Cheng Yang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; Shanghai GoBroad Cancer Hospital, China Pharmaceutical University, Shanghai, 200131, PR China.
| | - Chengjun Sui
- Department of Special Treatment, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, PR China.
| |
Collapse
|
43
|
Galassi C, Esteller M, Vitale I, Galluzzi L. Epigenetic control of immunoevasion in cancer stem cells. Trends Cancer 2024; 10:1052-1071. [PMID: 39244477 DOI: 10.1016/j.trecan.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024]
Abstract
Cancer stem cells (CSCs) are a poorly differentiated population of malignant cells that (at least in some neoplasms) is responsible for tumor progression, resistance to therapy, and disease relapse. According to a widely accepted model, all stages of cancer progression involve the ability of neoplastic cells to evade recognition or elimination by the host immune system. In line with this notion, CSCs are not only able to cope with environmental and therapy-elicited stress better than their more differentiated counterparts but also appear to better evade tumor-targeting immune responses. We summarize epigenetic modifications of DNA and histones through which CSCs evade immune recognition or elimination, and propose that such alterations constitute promising therapeutic targets to increase the sensitivity of some malignancies to immunotherapy.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
44
|
Luo S, Yue M, Wang D, Lu Y, Wu Q, Jiang J. Breaking the barrier: Epigenetic strategies to combat platinum resistance in colorectal cancer. Drug Resist Updat 2024; 77:101152. [PMID: 39369466 DOI: 10.1016/j.drup.2024.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/22/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. Platinum-based drugs, such as cisplatin and oxaliplatin, are frontline chemotherapy for CRC, effective in both monotherapy and combination regimens. However, the clinical efficacy of these treatments is often undermined by the development of drug resistance, a significant obstacle in cancer therapy. In recent years, epigenetic alterations have been recognized as key players in the acquisition of resistance to platinum drugs. Targeting these dysregulated epigenetic mechanisms with small molecules represents a promising therapeutic strategy. This review explores the complex relationship between epigenetic changes and platinum resistance in CRC, highlighting current epigenetic therapies and their effectiveness in countering resistance mechanisms. By elucidating the epigenetic underpinnings of platinum resistance, this review aims to contribute to ongoing efforts to improve treatment outcomes for CRC patients.
Collapse
Affiliation(s)
- Shiwen Luo
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ming Yue
- Department of Pharmacy, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Dequan Wang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yukang Lu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qingming Wu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China; Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Jue Jiang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
45
|
Wang K, Zhu S, Zhang Y, Wang Y, Bian Z, Lu Y, Shao Q, Jin X, Xu X, Mo R. Targeting the GTPase RAN by liposome delivery for tackling cancer stemness-emanated therapeutic resistance. J Control Release 2024; 375:589-600. [PMID: 39245420 DOI: 10.1016/j.jconrel.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Cancer therapeutic resistance as a common hallmark of cancer is often responsible for treatment failure and poor patient survival. Cancer stem-like cells (CSCs) are one of the main contributors to therapeutic resistance, cancer relapse and metastasis. Through screening from our in-house library of natural products, we found polyphyllin II (PPII) as a potent anti-CSC compound for triple-negative breast cancer (TNBC). To enhance anti-CSC selectivity and improve druggability of PPII, we leverage the liposome-mediated delivery technique for increasing solubility of PPII, and more significantly, attaining broader therapeutic window. Liposomal PPII demonstrates its marked potency to inhibit tumor growth, post-surgical recurrence and metastasis compared to commercial liposomal chemotherapeutics in the mouse models of CSC-enriched TNBC tumor. We further identify PPII as an inhibitor of the Ras-related nuclear (RAN) protein whose upregulated expression is correlated with poor clinical outcomes. The direct binding of PPII to RAN reduces TNBC stemness, thereby suppressing tumor progression. Our work offers a significance from drug discovery to drug delivery benefiting from liposome technique for targeted treatment of high-stemness tumor.
Collapse
Affiliation(s)
- Kaili Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Sitong Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yuqian Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Zhenqian Bian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yougong Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Quanlin Shao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Xiang Jin
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310032, China
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; Department of Pharmacy, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Center for Innovative Traditional Chinese Medicine Target and New Drug Research, International Institutes of Medicine, Zhejiang University, Yiwu 322001, Zhejiang, China.
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
46
|
Zhang T, Lei J, Zheng M, Wen Z, Zhou J. Nitric oxide facilitates the S-nitrosylation and deubiquitination of Notch1 protein to maintain cancer stem cells in human NSCLC. J Cell Mol Med 2024; 28:e70203. [PMID: 39523215 PMCID: PMC11550923 DOI: 10.1111/jcmm.70203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/19/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality, with tumour heterogeneity, fueled by cancer stem cells (CSCs), intricately linked to treatment resistance. Therefore, it is imperative to advance therapeutic strategies targeting CSCs in NSCLC. In this study, we utilized RNA sequencing to investigate metabolic pathway alterations in NSCLC CSCs and identified a crucial role of nitric oxide (NO) metabolism in governing CSC stemness, primarily through modulation of the Notch1 protein. Mechanistically, NO-induced S-nitrosylation of Notch1 facilitated its interaction with the deubiquitylase UCHL1, leading to increased Notch1 protein stability and enhanced CSC stemness. By inhibiting NO synthesis and downregulating UCHL1 expression, we validated the impact of NO on the Notch signalling pathway and CSC stemness. Importantly, targeting NO effectively reduced CSC populations within patient-derived organoids (PDOs) during radiotherapy. This mechanism presents a promising therapeutic target to surmount radiotherapy resistance in NSCLC treatment.
Collapse
Affiliation(s)
- Tenglong Zhang
- Department of Radiation OncologyThe First Affiliated Hospital of Soochow University, Soochow UniversitySuzhouChina
- Department of OncologyQingdao Municipal HospitalQingdaoChina
| | - Jiaxin Lei
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
| | - Ming Zheng
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
| | - Zhenke Wen
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
| | - Juying Zhou
- Department of Radiation OncologyThe First Affiliated Hospital of Soochow University, Soochow UniversitySuzhouChina
| |
Collapse
|
47
|
Al-Wahaibi LH, El-Sheref EM, Tawfeek HN, Abou-Zied HA, Rabea SM, Bräse S, Youssif BGM. Design, synthesis, and biological evaluation of novel quinoline-based EGFR/HER-2 dual-target inhibitors as potential anti-tumor agents. RSC Adv 2024; 14:32978-32991. [PMID: 39434991 PMCID: PMC11492426 DOI: 10.1039/d4ra06394e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
Dual targeting of EGFR and HER2 is a valid anti-cancer approach for treating solid tumors. We designed and synthesized a new series of EGFR/HER-2 dual-target inhibitors based on quinoline derivatives. The structure of the newly synthesized compounds was verified using 1H NMR, 13C NMR, and elemental analysis. The targeted compounds were tested for antiproliferative efficacy against four cancer cell lines. All the compounds had GI50s ranging from 25 to 82 nM, with breast (MCF-7) and lung (A-549) cancer cell lines being the most sensitive. Compound 5a demonstrated the most significant antiproliferative action. With inhibitory (IC50) values of 71 and 31 nM, respectively, compound 5a proved to be the most effective dual-target inhibitor of EGFR and HER-2, outperforming the reference erlotinib (IC50 = 80 nM) as an EGFR inhibitor but falling short of the clinically used agent lapatinib (IC50 = 26 nM) as a HER2 inhibitor. The apoptotic potential activity of 5a was examined, and the findings demonstrated that 5a promotes apoptosis by activating caspase-3, 8, and Bax while simultaneously reducing the expression of the anti-apoptotic protein Bcl-2. The docking studies provided valuable insights into the binding interactions of compounds 3e and 5a with EGFR, effectively rationalizing the observed SAR trends.
Collapse
Affiliation(s)
- Lamya H Al-Wahaibi
- Department of Chemistry, College of Sciences, Princess Nourah Bint Abdulrahman University Riyadh 11671 Saudi Arabia
| | - Essmat M El-Sheref
- Chemistry Department, Faculty of Science, Minia University El Minia 61519 Egypt
| | - Hendawy N Tawfeek
- Chemistry Department, Faculty of Science, Minia University El Minia 61519 Egypt
- Unit of Occupational of Safety and Health, Administration Office of Minia University El-Minia 61519 Egypt
| | - Hesham A Abou-Zied
- Medicinal Chemistry Department, Faculty of Pharmacy, Deraya University Minia Egypt
| | - Safwat M Rabea
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University Minia 61519 Egypt
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology Karlsruhe 76131 Germany
| | - Bahaa G M Youssif
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt +20-01098294419
| |
Collapse
|
48
|
Cheng X, Li P, Jiang R, Meng E, Wu H. ADC: a deadly killer of platinum resistant ovarian cancer. J Ovarian Res 2024; 17:196. [PMID: 39367438 PMCID: PMC11451100 DOI: 10.1186/s13048-024-01523-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024] Open
Abstract
Platinum is a key component of ovarian cancer systemic therapy. However, most patients will eventually face a recurrence, leading to chemotherapy resistance, especially against platinum. For individuals with platinum-resistant ovarian cancer (PROC), treatment options are limited, and their survival prospects are grim. The emergence of antibody-drug conjugates (ADCs) shows promises as a future treatment for PROC. This review synthesizes current research on the effectiveness of ADCs in treating PROC. It encapsulates the advancements and clinical trials of novel ADCs that target specific antigens such as Folate Receptor alpha (FRα), MUC16, NaPi2b, Mesothelin, Dipeptidase 3(DPEP3), and human epidermal growth factor receptor 2 (HER2), as well as tissue factor, highlighting their potential anti-tumor efficacy and used in combination with other therapies. The ADCs landscape in ovarian cancer therapeutics is swiftly evolving, promising more potent and efficacious treatment avenues.
Collapse
Affiliation(s)
- Xu Cheng
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China
| | - Ping Li
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China
| | - Rongqi Jiang
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China
| | - Enqing Meng
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China
| | - Hao Wu
- The Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China.
| |
Collapse
|
49
|
Erturk E, Ari F, Onur OE, Mustafa Gokgoz S, Tolunay S. Value of miR-31 and miR-150-3p as diagnostic and prognostic biomarkers for breast cancer. Mol Biol Rep 2024; 51:1030. [PMID: 39352561 DOI: 10.1007/s11033-024-09958-9] [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: 03/04/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND The most prevalent malignancy among women is breast cancer (BC). MicroRNAs (miRNAs) play a role in the initiation and progression of BC by influencing breast cancer stem cells (BCSCs) but the diagnostic and prognostic roles of those miRNAs on BC patients are still unknown. It was aimed to investigate expression profiles, diagnostic and prognostic potentials of BCSC-related miRNAs in different subtypes (Luminal A and B, HER2 + and TNBC) of BC patients. METHODS AND RESULTS Expression analysis of 15 BCSC-related miRNAs was performed in 50 breast tumor tissues and 20 adjacent non-tumor tissues obtained from BC patients using the qRT-PCR method. The expression levels of miR-31 and miR-150-3p were significantly upregulated in the tumor tissues compared to the adjacent non-tumor tissues (p < 0.05). miR-31 expression upregulated in the Luminal A and Luminal B group compared to non-tumor tissue (p < 0.05). miR-31 expression was determined to be significantly higher in the Luminal group (Luminal A and B) compared to the aggressive group (HER2 + and TNBC) (p < 0.05). According to the ROC analysis, the area under the curve (AUC) of miR-31 and miR-150-3p were 0.66 with a sensitivity of 68% and a specificity of 70%. A significant inverse correlation was observed between miR-31 expression with metastatic carcinoma status, in situ component, and Ki67 value in tumors, and high miR-150-3p expression was correlated with p63 expression (p < 0.05). CONCLUSION miR-31 and miR-150-3p have the potential to serve as biomarkers for guiding diagnosis, evaluating prognosis, and metastatic process in patients with BC.
Collapse
Affiliation(s)
- Elif Erturk
- Vocational School of Health Services, Bursa Uludag University, 16059, Bursa, Türkiye
| | - Ferda Ari
- Department of Biology, Science and Art Faculty, Bursa Uludag University, 16059, Bursa, Türkiye.
| | - Omer Enes Onur
- Department of Biology, Science and Art Faculty, Bursa Uludag University, 16059, Bursa, Türkiye
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Sehsuvar Mustafa Gokgoz
- Department of General Surgery, Faculty of Medicine, Bursa Uludag University, 16059, Bursa, Türkiye
| | - Sahsine Tolunay
- Department of Pathology, Faculty of Medicine, Bursa Uludag University, 16059, Bursa, Türkiye
| |
Collapse
|
50
|
Zheng D, Jin S, Liu PS, Ye J, Xie X. Targeting ferroptosis by natural products in pathophysiological conditions. Arch Toxicol 2024; 98:3191-3208. [PMID: 38987487 DOI: 10.1007/s00204-024-03812-4] [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: 05/09/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Ferroptosis is a form of cell death that is induced by iron-mediated accumulation of lipid peroxidation. The involvement of ferroptosis in different pathophysiological conditions has offered new perspectives on potential therapeutic interventions. Natural products, which are widely recognized for their significance in drug discovery and repurposing, have shown great promise in regulating ferroptosis by targeting various ferroptosis players. In this review, we discuss the regulatory mechanisms of ferroptosis and its implications in different pathological conditions. We dissect the interactions between natural products and ferroptosis in cancer, ischemia/reperfusion, neurodegenerative diseases, acute kidney injury, liver injury, and cardiomyopathy, with an emphasis on the relevance of ferroptosis players to disease targetability.
Collapse
Affiliation(s)
- Daheng Zheng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Shikai Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Pu-Ste Liu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Jianping Ye
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|