1
|
Li Y, Liu X, Dong Y, Zhou Y. Angiogenesis causes and vasculogenic mimicry formation in the context of cancer stem cells. Biochim Biophys Acta Rev Cancer 2025; 1880:189323. [PMID: 40239849 DOI: 10.1016/j.bbcan.2025.189323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
Tumor occurrence, development, invasion, and metastasis are regulated by multiple mechanisms. Among these, angiogenesis promotes tumor progression mainly by supplying tumor tissue and providing channels for tumor metastasis. Cancer stem cells (CSCs) are another important factor affecting tumor progression by involving in tumor initiation and development, while remaining insensitive to conventional antitumor treatments. Among treatment strategies for them, owing to the existence of alternative angiogenic pathways or the risk of damaging normal stem cells, the clinical effect is not ideal. Angiogenesis and CSCs may influence each other in this process. Tumor angiogenesis can support CSC self-renewal by providing a suitable microenvironment, whereas CSCs can regulate tumor neovascularization and mediate drug resistance to anti-angiogenic therapy. This review summarized the role of vascular niche formed by angiogenesis in CSC self-renewal and stemness maintenance, and the function of CSCs in endothelial progenitor cell differentiation and pro-angiogenic factor upregulation. We also elucidated the malignant loop between CSCs and angiogenesis promoting tumor progression. Additionally, we summarized and proposed therapeutic targets, including blocking tumor-derived endothelial differentiation, inhibiting pro-angiogenic factor upregulation, and directly targeting endothelial-like cells comprising CSCs. And we analyzed the feasibility of these strategies to identify more effective methods to improve tumor treatment.
Collapse
Affiliation(s)
- Ying Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaofang Liu
- Department of Anus and Intestine Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yaodong Dong
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Yingying Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
2
|
Shi Y, Cao Z, Ge L, Lei L, Tao D, Zhong J, Xu D, Geng T, Li X, Li Z, Xing S, Wu X, Wang Z, Li L. Rotenone adaptation promotes migration and invasion of p53-wild-type colon cancer through lipid metabolism. Clin Transl Oncol 2025; 27:2557-2567. [PMID: 39612123 DOI: 10.1007/s12094-024-03785-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/10/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The association between mitochondrial dysfunction and multiple metabolic adaptations is increasingly being proven. We previously elucidated that mitochondrial complex I deficiency can promote glycolysis in mut-p53 SW480 cells. However, studies have revealed a phenotype with attenuated glycolysis but enhanced fatty acid oxidation (FAO) in invasive tumors. The interplay between complex I and FAO in carcinogenesis remains obscure. METHODS The p53 wild-type RKO cells were exposed to rotenone over at least 2 months to acquire rotenone adaptation cells. Then the transwell invasion assays and expression of metabolic enzymes were first detected in rotenone adaptation cells to illustrate whether rotenone adaptation is correlated with the invasion and FAO. The levels of epithelial-to-mesenchymal transition (EMT)-related proteins and acetyl-CoA in rotenone adaptation cells treated with etomoxir (ETO) and acetate were evaluated to verify the role of CPT1A in regulating invasion. Finally, the levels of reactive oxygen species (ROS) were detected. Meanwhile, the invasiveness and histone acetylation levels of rotenone adaptation cells were observed after adding an ROS inhibitor (N-acetyl-L-cysteine NAC) to demonstrate the molecular connection between FAO and invasion during rotenone adaptation. RESULTS We found long-term exposure to rotenone (a mitochondrial complex I inhibitor) led to EMT and high CPT1A expression in wt-p53 colon cancer. The inhibition of CPT1A suppressed the invasion and reduced histone acetylation, which was rescued by supplementing with acetate. Mechanistically, ROS is crucial for lipid metabolism remodeling. CONCLUSION Our study provides a novel understanding of the role of complex I in lipid reprogramming facilitating colon cancer invasion and metastasis.
Collapse
Affiliation(s)
- Yingying Shi
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Zhen Cao
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Ling Ge
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Lin Lei
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Dan Tao
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Juan Zhong
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Dan Xu
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Tao Geng
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Xuetao Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Ziwei Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Shuaishuai Xing
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Xinyu Wu
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Zhongxu Wang
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Linjun Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China.
| |
Collapse
|
3
|
Huang X, Hou S, Li Y, Xu G, Xia N, Duan Z, Luo K, Tian B. Targeting lipid metabolism via nanomedicine: A prospective strategy for cancer therapy. Biomaterials 2025; 317:123022. [PMID: 39754967 DOI: 10.1016/j.biomaterials.2024.123022] [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: 09/09/2024] [Revised: 11/20/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025]
Abstract
Lipid metabolism has been increasingly recognized to play an influencing role in tumor initiation, progression, metastasis, and therapeutic drug resistance. Targeting lipid metabolic reprogramming represents a promising therapeutic strategy. Despite their structural complexity and poor targeting efficacy, lipid-metabolizing drugs, either used alone or in combination with chemotherapeutic agents, have been employed in clinical practice. The advent of nanotechnology offers new approaches to enhancing therapeutic effects, includingthe targeted delivery and integration of lipid metabolic reprogramming with chemotherapy, photodynamic therapy (PDT), and immunotherapy. The integrated nanoformulation, nanomedicine, could significantly advance the field of lipid metabolism therapy. In this review, we will briefly introduce the concept of cancer lipid metabolism reprogramming, then elaborate the latest advances in engineered nanomedicine for targeting lipid metabolism during cancer treatment, and finally provide our insights into future perspectives of nanomedicine for interference with lipid metabolism in the tumor microenvironment.
Collapse
Affiliation(s)
- Xing Huang
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shengzhong Hou
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinggang Li
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Xu
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Ning Xia
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.
| | - Kui Luo
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.
| | - Bole Tian
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Chen WM, Zhang XP, Sun X, Liu HC, Yan YY, Wei X, Liang Y, Feng Y, Chen Z, Jia Y, Jiang C, Yan Q, Li L. QSOX2-Mediated Disulfide Bond Modification Enhances Tumor Stemness and Chemoresistance by Activating TSC2/mTOR/c-Myc Feedback Loop in Esophageal Squamous Cell Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e00597. [PMID: 40433832 DOI: 10.1002/advs.202500597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/29/2025] [Indexed: 05/29/2025]
Abstract
Disulfide bond modification is critical in maintaining protein structure and activity, but its roles in regulating tumor stemness and chemoresistance remain underexplored. Here, Quiescin Sulfhydryl Oxidase 2 (QSOX2) is identified, a protein involved in disulfide bond formation, is highly expressed in esophageal squamous cell carcinoma (ESCC), and is associated with poor patient prognosis. Functional analyses demonstrated that QSOX2 overexpression markedly potentiated tumor stemness and further promoted chemoresistance, proliferation, and metastasis of ESCC cells. Mechanistically, QSOX2 enhances disulfide bond formation in TSC Complex Subunit 2 (TSC2), stabilizing TSC2-Akt interactions, facilitating phosphorylation of TSC2 at the Ser939 by Akt, and further activating mTOR/4E-BP1/c-Myc signaling axis. Intriguingly, cancer-associated fibroblasts-secreted IGF-1 upregulates QSOX2 expression via IGF1R/Akt/mTOR/c-Myc pathway, establishing a positive feedback loop that sustains ESCC cell stemness. Targeting QSOX2 with Ebselen, in combination with mTOR inhibitor Rapamycin and chemotherapy, effectively downregulates c-Myc expression and induces tumor dormancy in a mouse xenograft model. Therefore, the findings reveal that QSOX2-mediated disulfide bond modification enhances tumor stemness by activating mTOR signaling, highlighting a promising therapeutic target in ESCC.
Collapse
Affiliation(s)
- Wo-Ming Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| | - Xiao-Ping Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| | - Xiao Sun
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| | - Hai-Cheng Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, P. R. China
| | - Yuan-Yuan Yan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| | - Xue Wei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| | - Yu Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| | - Yue Feng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| | - Zhengjie Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| | - Yongxu Jia
- Department of Clinical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 451191, P. R. China
- State Key Laboratory of Metabolic Desregulation & Prevention and Treatment of Esphageal Cancer, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 451191, P. R. China
| | - Chen Jiang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Qian Yan
- Guangdong Institute of Gastroenterology and Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, P. R. China
| | - Lei Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| |
Collapse
|
5
|
Yang Y, Deng S, Yin Q, Zhang T, Liu T, Wu D, Xu Y. RUNX1 promotes colorectal cancer progression by activating SERPINE1 transcription. Biochem Biophys Res Commun 2025; 760:151732. [PMID: 40164015 DOI: 10.1016/j.bbrc.2025.151732] [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: 01/17/2025] [Revised: 03/04/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is among the most prevalent malignancies affecting the digestive system globally, considerably endangering public health. Runt-related transcription factor 1 (RUNX1) is a key regulator that influences the progression of cancer. In this study, we investigated the involvement of RUNX1 in CRC progression and its underlying mechanisms. METHODS The expression levels and prognostic significance of RUNX1 in CRC were analyzed based on data from the TNMplot, Gene Expression Profiling Interactive Analysis, and Gene Expression Omnibus databases as well as tissue microarrays. RUNX1 was knocked down or overexpressed in HT29 and HCT116 cells. The proliferation, migration, and invasion of CRC cells were evaluated. The target gene of RUNX1 was identified using PCR array analysis and validated via chromatin immunoprecipitation and luciferase reporter assays. The effects of RUNX1 on CRC cells were verified in vivo. RESULTS RUNX1 expression in CRC tissues was significantly higher than that in adjacent non-tumorous tissues and was associated with an unfavorable prognosis. Silencing RUNX1 significantly suppressed the proliferation, migratory capability, and invasive potential of CRC cells; overexpressing RUNX1 had the opposite effect. SERPINE1 was identified as a direct transcriptional target of RUNX1. In vivo experiments further validated that RUNX1-promoted CRC tumor growth. Mechanistically, RUNX1 promotes CRC progression by promoting the transcription of SERPINE1. CONCLUSIONS Our findings revealed that RUNX1 promotes CRC progression by activating SERPINE1 transcription, suggesting that RUNX1 is a potential prognostic biomarker and therapeutic target in CRC.
Collapse
Affiliation(s)
- Yueyan Yang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Shihua Deng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Qing Yin
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Ting Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Teng Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Dongming Wu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China.
| | - Ying Xu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; Sichuan Clinical Research Center for Radiation and Therapy, The First Affiliated Hospital of Chengdu Medical College, 610500, PR China.
| |
Collapse
|
6
|
Wang W, Zhang Y. Analysis of the Predictive Efficacy and Influencing Factors of Serum Tie-1, FoxO3a, and PKD1 for Lymph Node Metastasis in Cervical Cancer. Int J Womens Health 2025; 17:1215-1224. [PMID: 40331131 PMCID: PMC12053925 DOI: 10.2147/ijwh.s512411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/05/2025] [Indexed: 05/08/2025] Open
Abstract
Objective To investigate the factors affecting lymph node metastasis (LNM) in patients with cervical cancer and the predictive efficacy of serum tyrosine kinase receptor 1 (Tie-1), serum Forkhead Framing Protein O3a (FoxO3a), and protein kinase D1 (PKD1). Methods Cervical cancer patients were categorized into 60 cases of LNM-positive group and 320 cases of LNM-negative group according to whether LNM occurred or not. The levels of serum Tie-1, FoxO3a and PKD1 were tested. Multivariate logistic regression analysis was conducted to identify the risk factors for cervical cancer induced lymph node metastasis (LNM). Receiver operating characteristic (ROC) curves were plotted to analyze the predictive value of various indicators for LNM in cervical cancer. Results The percentage of patients with FIGO stage IIa, combined paracervical infiltration and myometrial infiltration was significantly higher in the LNM-positive group than in the LNM-negative group (P<0.05). Huanz serum levels of Tie-1 and PKD1 in the LNM-positive group were significantly higher than those in the LNM-negative group, and the relative expression of FoxO3a was significantly lower than that in the LNM-negative group (P<0.05). The results of logistic regression analysis showed that FIGO stage, parietal infiltration, myometrial infiltration, serum Tie-1, PKD1 were LNM-positive in cervical cancer patients (P<0.05), and low level of relative expression of serum FoxO3a was a protective factor (P<0.05). The cutoff of serum Tie-1, FoxO3a, and PKD1 levels for predicting the occurrence of LNM in cervical cancer were 1.97 ng/mL, 0.54, and 113.26 μg/L, and the area under the ROC curve (AUC) was 0.852, 0.827, 0.844, respectively. Conclusion Serum Tie-1, FoxO3a and PKD1 have certain predictive efficacy for lymph node metastasis, and the combination of these tests can improve the predictive accuracy.
Collapse
Affiliation(s)
- Wei Wang
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, 110000, People’s Republic of China
| | - Yi Zhang
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, 110000, People’s Republic of China
| |
Collapse
|
7
|
Ping P, Ma Y, Xu X, Li J. Reprogramming of fatty acid metabolism in thyroid cancer: Potential targets and mechanisms. Chin J Cancer Res 2025; 37:227-249. [PMID: 40353071 PMCID: PMC12062987 DOI: 10.21147/j.issn.1000-9604.2025.02.09] [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: 01/16/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Thyroid cancer (TC) is one of the most common endocrine system tumors, and its incidence continues to increase worldwide. Although most TC patients have a good prognosis, especially with continuous advancements in surgery, radioactive iodine therapy, chemotherapy, endocrine therapy and targeted therapy, the effectiveness of disease treatment has significantly improved. However, there are still some cases with a higher risk of death and greater aggressiveness. In these more challenging advanced or highly aggressive cases, tyrosine kinase inhibitors appear to be an effective treatment option. Unfortunately, these drugs are less than ideal in terms of efficacy because of their toxicity and potential for intrinsic or acquired resistance. Therefore, exploring new strategies targeting the metabolic characteristics of TC cells and overcoming drug resistance barriers in existing treatments have become key topics in the current field of TC research. In recent years, lipid metabolic reprogramming has gained attention as an important aspect of cancer development. Lipid metabolic reprogramming not only participates in the formation of the cell membrane structure, but also plays an important role in signal transduction and promoting cell proliferation. In particular, fatty acid (FA) metabolic reprogramming has attracted widespread attention and plays an important role in multiple aspects such as tumor growth, metastasis, enhanced invasive ability, immune escape, and drug resistance. Although TC is considered a disease that is highly dependent on specific types of metabolic activities, a comprehensive understanding of the specific mechanism of action of FA metabolic reprogramming in this process is lacking. This article aims to review how FA metabolic reprogramming participates in the occurrence and development of TC, focusing on the impact of abnormal FA metabolic pathways and changes in the expression and regulation of related genes over the course of this disease. By examining the complex interactions between FA metabolic disorders and carcinogenic signaling pathways in depth, we aim to identify new therapeutic targets and develop more precise and effective treatments for TC.
Collapse
Affiliation(s)
- Pengbin Ping
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
- Department of Radiation Therapy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Yuhong Ma
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Xiaoying Xu
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Juan Li
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
8
|
Qin X, Guo J, Li H, He H, Cai F, Chen X, Chen M, Chen T, Ma L. Selenium Electrophilic Center Responsive to Biological Electron Donors for Efficient Chemotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412062. [PMID: 39950936 PMCID: PMC11984860 DOI: 10.1002/advs.202412062] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/24/2024] [Indexed: 04/12/2025]
Abstract
Designing drugs to intelligently respond to different ratio of biological electron donors/receptors in cancer cells and normal cells is a promising strategy to achieve highly effective and less toxic chemotherapy. Herein by employing metal center to active the selenium-containing electrophilic center drug Ru(phtpy-NO2)(phenSe)Cl (RuSe) with strongly polarization characteristics are synthesized which can efficiently shuttle electrons from biological electron donors to convert to oxidative stress. The rate of electron transfer at the selenium electrophilic center is 1.81 times higher in cancer cell environments compared to normal cell environments. This results in the selenium electrophilic center being 14.98 times more lethal to cancer cells than to normal cells. Experimental results demonstrate that the transport of electrons process is carried out via selenium radicals intermediate and the rate of electron transport is positively correlated with the polarization properties of the electrophilic center atoms. The selenium electrophilic center transports bioactive electrons to generate a large number of superoxide anions leading to DNA damage and a decrease in mitochondrial membrane potential which further activates the p53 signaling pathway and amplifies the cancer cell-killing effect after transporting bioactive electrons. This work provides a new avenue for the design of efficient and less toxic chemotherapeutic agents.
Collapse
Affiliation(s)
- Xiaoyu Qin
- Department of Pharmacy and General Surgery of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionJinan UniversityGuangzhou510632China
| | - Junxian Guo
- Department of Pharmacy and General Surgery of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionJinan UniversityGuangzhou510632China
| | - Hui Li
- Department of Pharmacy and General Surgery of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionJinan UniversityGuangzhou510632China
| | - Hanlong He
- Department of Pharmacy and General Surgery of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionJinan UniversityGuangzhou510632China
| | - Fei Cai
- Department of Pharmacy and General Surgery of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionJinan UniversityGuangzhou510632China
| | - Xinyan Chen
- Department of Pharmacy and General Surgery of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionJinan UniversityGuangzhou510632China
| | - Mingkai Chen
- Department of Pharmacy and General Surgery of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionJinan UniversityGuangzhou510632China
| | - Tianfeng Chen
- Department of Pharmacy and General Surgery of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionJinan UniversityGuangzhou510632China
| | - Li Ma
- Department of Pharmacy and General Surgery of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionJinan UniversityGuangzhou510632China
| |
Collapse
|
9
|
Pan Y, Yuan C, Zeng C, Sun C, Xia L, Wang G, Chen X, Zhang B, Liu J, Ding ZY. Cancer stem cells and niches: challenges in immunotherapy resistance. Mol Cancer 2025; 24:52. [PMID: 39994696 PMCID: PMC11852583 DOI: 10.1186/s12943-025-02265-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
Cancer stem cells (CSCs) are central to tumor progression, metastasis, immune evasion, and therapeutic resistance. Characterized by remarkable self-renewal and adaptability, CSCs can transition dynamically between stem-like and differentiated states in response to external stimuli, a process termed "CSC plasticity." This adaptability underpins their resilience to therapies, including immune checkpoint inhibitors and adoptive cell therapies (ACT). Beyond intrinsic properties, CSCs reside in a specialized microenvironment-the CSC niche-which provides immune-privileged protection, sustains their stemness, and fosters immune suppression. This review highlights the critical role of CSCs and their niche in driving immunotherapy resistance, emphasizing the need for integrative approaches to overcome these challenges.
Collapse
Affiliation(s)
- Yonglong Pan
- Hepatic Surgery Center, Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Cellular Signaling laboratory, Key laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chaoyi Yuan
- Hepatic Surgery Center, Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chenglong Zeng
- Hepatic Surgery Center, Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chaoyang Sun
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center, Key Laboratory of the MOE, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Limin Xia
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guihua Wang
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Tongji Hospital, GI Cancer Research Institute, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission, Chinese Academy of Medical Sciences, Wuhan, 430030, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jianfeng Liu
- Cellular Signaling laboratory, Key laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Ze-Yang Ding
- Hepatic Surgery Center, Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
10
|
Zhang C, Yang T, Yu Y, Jia Q, Xiao WM, Liu S, Yu ZH, Wen CL, Wei Y, Li H, Lü MH. Causal roles of immune cells and metabolites in chronic pancreatitis: a mendelian randomization study. Hereditas 2025; 162:20. [PMID: 39940040 PMCID: PMC11816568 DOI: 10.1186/s41065-025-00378-8] [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: 11/14/2024] [Accepted: 01/26/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Previous research has established a correlation between immune cells and an increased likelihood of Chronic pancreatitis (CP). However, studies investigating the causal relationship remain limited. METHODS This study utilized publicly available genome-wide association study (GWAS) databases and conducted a two-sample Mendelian randomization (MR) analysis to examine the causal relationships (CRs) among 731 immune cells, 1,400 metabolites, and CP. Mediation MR analysis was also performed to assess whether metabolites serve as mediators in the relationship between immune cells and CP. RESULTS Our study identified four immune cell types that act as risk factors for CP, with odds ratios (OR) ranging between 1.076 and 1.177. In contrast, three immune cell types were found to serve as protective factors, exhibiting OR values between 0.846 and 0.913. Additionally, four metabolites were implicated as risk factors for CP, with OR values ranging from 1.243 to 1.334. On the other hand, eight metabolites were discovered to have a protective effect, with OR values between 0.580 and 0.871. Mediation analysis revealed that cholesterol levels mediate the causal relationship between immune cell cells and CP, with a mediation effect of 0.00918, accounting for 9.18% of the total effect. CONCLUSIONS Our findings provide valuable insights into the genetic underpinnings of CP, highlighting the role of immune cells and plasma metabolites in its pathogenesis. The mediation analysis further suggests that the presence of CD25 on IgD-CD38-B cells may facilitate CP development through the elevation of cholesterol levels. These results not only deepen our understanding of CP but also suggest potential biological targets for therapeutic intervention. Future clinical research should focus on these mediators to develop more effective treatment strategies for CP.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Tao Yang
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yuan Yu
- Gulin County People's Hospital, Luzhou, Sichuan Province, China
| | - Qian Jia
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Wan-Meng Xiao
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Sha Liu
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Ze-Hui Yu
- Laboratory Animal Center, Southwest Medical University, Luzhou, Sichuan, China
- Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Cheng-Li Wen
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yan Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Hao Li
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| | - Mu-Han Lü
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|