1
|
Chen S, Cheng J, Liu S, Shan D, Wang T, Wang X. Urinary exosomal lnc-TAF12-2:1 promotes bladder cancer progression through the miR-7847-3p/ASB12 regulatory axis. Genes Dis 2025; 12:101384. [PMID: 40297540 PMCID: PMC12036056 DOI: 10.1016/j.gendis.2024.101384] [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: 11/10/2023] [Revised: 06/02/2024] [Accepted: 06/22/2024] [Indexed: 04/30/2025] Open
Abstract
Exosomes encompass a great deal of valuable biological information and play a critical role in tumor development. However, the mechanism of exosomal lncRNAs remains poorly elucidated in bladder cancer (BCa). In this study, we identified exosomal lnc-TAF12-2:1 as a novel biomarker in BCa diagnosis and aimed to investigate the underlying biological function. Dual luciferase reporter assay, RNA immunoprecipitation (RIP), RNA pulldown assays, and xenograft mouse model were used to verify the competitive endogenous RNA mechanism of lnc-TAF12-2:1. We found exosomal lnc-TAF12-2:1 up-regulated in urinary exosomes, tumor tissues of patients, and BCa cells. Down-regulation of lnc-TAF12-2:1 impaired BCa cell proliferation and migration, and promoted cell cycle arrest at the G0/G1 phase and cell apoptosis. The opposite effects were also observed when lnc-TAF12-2:1 was overexpressed. lnc-TAF12-2:1 was transferred by intercellular exosomes to modulate malignant biological behavior. Mechanistically, lnc-TAF12-2:1 packaged in the exosomes relieved the miRNA-mediated silence effect on ASB12 via serving as a sponger of miR-7847-3p to accelerate progression in BCa. ASB12 was also first proved as an oncogene to promote cell proliferation and migration and depress cell cycle arrest and cell apoptosis in our data. In conclusion, exosomal lnc-TAF12-2:1, located in the cytoplasm of BCa, might act as a competitive endogenous RNA to competitively bind to miR-7847-3p, and then be involved in miR-7847-3p/ASB12 regulatory axis to promote tumorigenesis, which provided a deeper insight into the molecular mechanism of BCa.
Collapse
Affiliation(s)
- Song Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Institute of Urology, Wuhan University, Wuhan, Hubei 430071, China
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, Hubei 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, Hubei 430071, China
| | - Jie Cheng
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Shuangtai Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Institute of Urology, Wuhan University, Wuhan, Hubei 430071, China
| | - Danni Shan
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, Hubei 430071, China
| | - Ting Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Institute of Urology, Wuhan University, Wuhan, Hubei 430071, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Institute of Urology, Wuhan University, Wuhan, Hubei 430071, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, Hubei 430071, China
- Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, Hubei 430071, China
- TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
2
|
Yang X, Wu X, Hao X, Li T, Guo H, Yang R. Unleashing the therapeutic potential of tumor-draining lymph nodes: spotlight on bladder cancer. J Transl Med 2025; 23:489. [PMID: 40301883 PMCID: PMC12042586 DOI: 10.1186/s12967-024-05864-7] [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: 08/30/2024] [Accepted: 11/07/2024] [Indexed: 05/01/2025] Open
Abstract
Tumor-draining lymph nodes (TDLNs) are often involved during the metastasis of bladder cancer (BC), which is associated with a poor prognosis. Recent studies have shown that TDLNs are a major source of host anti-tumor immunity, which can impede tumor progression and favor tumor immunotherapy. However, during tumor progression, various tumor-derived mediators modulate the TDLN microenvironment, impairing their protective function. Ultimately, TDLNs provide the soil for the proliferation and dissemination of tumor cells. Therefore, surgical removal of TDLNs is commonly recommended in various solid tumors to prevent metastasis, but this poses significant challenges for leveraging TDLNs in immunotherapy. Additionally, lymph node dissection (LND) has not shown survival benefits in some tumors. Hence, the decision to remove TDLNs in oncological treatment needs to be reconsidered. Herein, we spotlight the TDLNs of BC and introduce how BC cells modulate stromal cells and immune cells to shape an immunosuppressive TDLN microenvironment for BC progression. We summarize the existing therapeutic strategies to reinvigorate anti-tumor immunity in TDLNs. Furthermore, we discuss whether to preserve TDLNs and the role of LND during oncological treatment.
Collapse
Affiliation(s)
- Xin Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiangyu Wu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuyang Hao
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tianhang Li
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China.
- Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
3
|
Cao LM, Qiu YZ, Li ZZ, Wang GR, Xiao Y, Luo HY, Liu B, Wu Q, Bu LL. Extracellular Vesicles: Hermes between cancers and lymph nodes. Cancer Lett 2025; 623:217735. [PMID: 40268131 DOI: 10.1016/j.canlet.2025.217735] [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: 02/05/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
Cancer is one of the main causes of death and a major obstacle to increasing life expectancy in all countries of the world. Lymph node metastasis (LNM) of in cancer patients indicates poor prognosis and it is an important indication to determine the therapeutic regime. Therefore, more attention should be given to the molecular mechanics of tumor lymphangiogenesis and LNM. Extracellular vesicles (EVs) are nanoscale cargo-bearing membrane vesicles that can serve as key mediators for the intercellular communication. Like Hermes, the messenger of the Greek gods, EVs can be secreted by tumor cells to regulate the LNM process. Many evidence has proved the clinical correlation between EVs and LNM in various cancer types. EVs plays an active role in the process of metastasis by expressing its connotative molecules, including proteins, nucleic acids, and metabolites. However, the clear role of EVs in the process of cancer LNM has not been thoroughly studied yet. In this review, we will summarize the clinical and mechanical findings of EVs regulating role on cancer LNM, and discuss the advanced modification of the research proposal. We propose the "PUMP" principle of EVs in LNM, including Preparation, Unleash, Migration, and Planting.
Collapse
Affiliation(s)
- Lei-Ming Cao
- 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, 430079, China
| | - Yu-Zhong Qiu
- 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, 430079, China
| | - Zi-Zhan Li
- 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, 430079, 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, 430079, 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, 430079, China
| | - Han-Yue Luo
- 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, 430079, 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, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan, 430071, 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, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
4
|
Yu Z, Fu J, Mantareva V, Blažević I, Wu Y, Wen D, Battulga T, Wang Y, Zhang J. The role of tumor-derived exosomal LncRNA in tumor metastasis. Cancer Gene Ther 2025; 32:273-285. [PMID: 40011710 DOI: 10.1038/s41417-024-00852-x] [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: 04/24/2024] [Revised: 10/22/2024] [Accepted: 11/05/2024] [Indexed: 02/28/2025]
Abstract
Tumor metastasis regulated by multiple complicated pathways is closely related to variations in the tumor microenvironment. Exosomes can regulate the tumor microenvironment through various mechanisms. Exosomes derived from tumor cells carry a variety of substances, including long non-coding RNAs (lncRNAs), play important roles in intercellular communication and act as critical determinants influencing tumor metastasis. In this review, we elaborate on several pivotal processes through which lncRNAs regulate tumor metastasis, including the regulation of epithelial‒mesenchymal transition, promotion of angiogenesis and lymphangiogenesis, enhancement of the stemness of tumor cells, and evasion of immune clearance. Additionally, we comprehensively summarized a diverse array of potential tumor-derived exosomal lncRNA biomarkers to facilitate accurate diagnosis and prognosis in a clinical setting.
Collapse
Affiliation(s)
- Zhile Yu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Jiali Fu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Vanya Mantareva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Bld. 9, 1113, Sofia, Bulgaria
| | - Ivica Blažević
- Department of Organic Chemistry, Faculty of Chemistry and Technology, University of Split, Ruđera Boškovića 35, 21000, Split, Croatia
| | - Yusong Wu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Dianchang Wen
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Tungalag Battulga
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia.
| | - Yuqing Wang
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China.
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 510140, PR China.
| | - Jianye Zhang
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China.
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, 511518, PR China.
| |
Collapse
|
5
|
Wan X, Wang D, Zhang X, Xu M, Huang Y, Qin W, Chen S. Unleashing the power of urine‑based biomarkers in diagnosis, prognosis and monitoring of bladder cancer (Review). Int J Oncol 2025; 66:18. [PMID: 39917986 PMCID: PMC11837902 DOI: 10.3892/ijo.2025.5724] [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: 09/23/2024] [Accepted: 01/13/2025] [Indexed: 02/21/2025] Open
Abstract
Bladder cancer (BCa) is a prevalent malignant neoplasm of the urinary tract with high incidence rate, frequent recurrence and rapid disease progression. Conventional approaches for diagnosing, prognosticating and monitoring BCa often rely on invasive procedures such as cystoscopy and tissue biopsy, which are associated with high costs and low patient compliance for follow‑up. Liquid biopsies have advantages, such as being non‑invasive, real‑time, and reproducible, in obtaining diverse biomarkers derived from cellular, molecular, proteomic and genetic signatures in urine or plasma samples. Although plasma‑based biomarkers have been clinically validated, urine provides greater specificity for directly assessing biological materials from urological sources. The present review summarizes advancements and current limitations in urinary protein, genetic and epigenetic biomarkers for disease progression and treatment response of BC, compares performance and application scenarios of urine and blood biomarkers and explores how urinary biomarkers may serve as an alternative or complementary tool to traditional diagnostic methods. The integration of urine‑based or plasma‑based biomarkers into existing diagnostic workflows offers promising avenues for improving accuracy and efficiency of diagnosis in the management of BCa. Notably, the emergence of synthetic biomarkers and urine metabolites, combined with artificial intelligence or bioinformatic technologies, has promise in the screening of potential targets. Continued research and validation efforts are needed to translate these findings into routine clinical practice, ultimately improving patient outcomes and decreasing the burden of BCa.
Collapse
Affiliation(s)
- Xuebin Wan
- Department of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
- Department of Research and Development, HaploX Biotechnology, Co., Ltd., Shenzhen, Guangdong 518057, P.R. China
| | - Dan Wang
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and Genetics, University of Goettingen, Göttingen D-37077, Germany
| | - Xiaoni Zhang
- Department of Research and Development, HaploX Biotechnology, Co., Ltd., Shenzhen, Guangdong 518057, P.R. China
| | - Mingyan Xu
- Department of Research and Development, HaploX Biotechnology, Co., Ltd., Shenzhen, Guangdong 518057, P.R. China
| | - Yuying Huang
- Department of Pediatrics, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Wenjian Qin
- Department of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
| | - Shifu Chen
- Department of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
- Department of Research and Development, HaploX Biotechnology, Co., Ltd., Shenzhen, Guangdong 518057, P.R. China
| |
Collapse
|
6
|
Dou X, Feng C, Li J, Jiang E, Shang Z. Extracellular vesicle-mediated crosstalk in tumor microenvironment dominates tumor fate. Trends Cell Biol 2025; 35:230-247. [PMID: 39327161 DOI: 10.1016/j.tcb.2024.08.008] [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: 03/15/2024] [Revised: 08/16/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
The tumor microenvironment (TME) is a complex and heterogeneous system containing various cells cooperating and competing with each other. Extracellular vesicles (EVs) differing in form and content are important intercellular communication mediators in the TME. Previous studies have focused on the cargoes within EVs rather than on the donors from which they originate and the recipient cells that exert their effects. Therefore, we provide here a detailed overview of the important roles of EVs in shaping tumor fate, highlighting their various mechanisms of intercellular dialog within the TME. We evaluate recent advances and also raise unresolved challenges to provide new ideas for clinical treatment strategies using EVs.
Collapse
Affiliation(s)
- Xinyu Dou
- 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 430079, China
| | - Chunyu Feng
- 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 430079, China
| | - Ji Li
- 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 430079, China
| | - Erhui Jiang
- 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 430079, China; Department of Oral and Maxillofacial Head Neck Surgery, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan 430079, China.
| | - Zhengjun Shang
- 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 430079, China; Department of Oral and Maxillofacial Head Neck Surgery, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan 430079, China.
| |
Collapse
|
7
|
Saadh MJ, Allela OQB, Kareem RA, Ballal S, Chahar M, Saini S, Prasad GVS, Sameer HN, Hamad AK, Athab ZH, Adil M. The role of exosomal non-coding RNAs in the breast cancer tumor microenvironment. Funct Integr Genomics 2025; 25:32. [PMID: 39891771 DOI: 10.1007/s10142-025-01531-2] [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: 10/07/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
The leading form of cancer affecting females globally is breast cancer, characterized by an unregulated growth of cells within the breast. Therefore, examining breast tissue is crucial in accurately identifying and treating this disease. Exosomes are very small enclosures bounded by a layer of cells and produced by a variety of cells present in the cancerous tissue surroundings. They play a crucial role in several biological functions in cancerous tumors. These exosomes carry non-coding RNAs (ncRNAs) and are discharged into the TME, where they are instrumental in the development and advancement of tumors. Additionally, the ncRNAs enclosed in exosomes act as significant mediators of communication within cells. Consequently, there is limited comprehension regarding the precise roles and targets of exosomal RNA in regulation, as research in this area is still in its preliminary phases. This piece provides a comprehensive overview of the latest studies on exosomes, delving into their impact on the behavior of cancer cells and immune cells. Moreover, it presents a compilation of the diverse forms of non-coding RNA molecules found in exosomes released by both cancerous and supportive cells, including circular RNAs, microRNAs, and long non-coding RNAs. Current research has proven the noteworthy influence that non-coding RNA molecules have on the progression, proliferation, drug resistance, and immune responses of breast cancer cells.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, 11831, Amman, Jordan
| | | | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Suman Saini
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, 140307, Mohali, Punjab, India
| | - G V Siva Prasad
- Department of Basic Sciences and Humanities, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, 64001, Dhi Qar, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy college, Al-Farahidi University, 00964, Baghdad, Iraq
| |
Collapse
|
8
|
Ning W, Yang J, Ni R, Yin Q, Zhang M, Zhang F, Yang Y, Zhang Y, Cao M, Jin L, Pan Y. Hypoxia induced cellular and exosomal RPPH1 promotes breast cancer angiogenesis and metastasis through stabilizing the IGF2BP2/FGFR2 axis. Oncogene 2025; 44:147-164. [PMID: 39496940 DOI: 10.1038/s41388-024-03213-y] [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: 08/20/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024]
Abstract
Metastasis is the major cause of breast cancer mortality, with angiogenesis and tumor-released exosomes playing key roles. However, the communication between breast cancer cells and endothelial cells and its role in tumor metastasis remains unclear. Here, we characterize a long noncoding RNA, RPPH1, which is upregulated in breast cancer tissues and positively associated with poor prognosis. Hypoxia microenvironment upregulates the expression of RPPH1 in breast cancer cells, and promotes its packaging into exosomes through hnRNPA1, leading to the maintenance of stemness and aggressive traits in cancer cells and angiogenesis in endothelial cells. The function of cellular and exosomal RPPH1 was confirmed in the MMTV-PyMT mouse model, in which ASO-RPPH1 therapy effectively inhibited tumor progression and metastasis. Mechanistically, RPPH1 protects IGF2BP2 from ubiquitination-induced degradation, stabilizes N6-methyladenosine (m6A)-modified FGFR2 mRNA, and activates the PI3K/AKT pathway. Our research unveils the role of RPPH1 in breast cancer metastasis and highlights its potential as a therapeutic target.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/blood supply
- Female
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Animals
- Exosomes/metabolism
- Exosomes/genetics
- Mice
- RNA-Binding Proteins/metabolism
- RNA-Binding Proteins/genetics
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Neoplasm Metastasis
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Signal Transduction
- Cell Hypoxia
- Tumor Microenvironment
- Angiogenesis
Collapse
Affiliation(s)
- Wentao Ning
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jingyan Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ruiqi Ni
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qianqian Yin
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Manqi Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fangfang Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yue Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Meng Cao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
9
|
Shirani N, Abdi N, Chehelgerdi M, Yaghoobi H, Chehelgerdi M. Investigating the role of exosomal long non-coding RNAs in drug resistance within female reproductive system cancers. Front Cell Dev Biol 2025; 13:1485422. [PMID: 39925739 PMCID: PMC11802832 DOI: 10.3389/fcell.2025.1485422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Exosomes, as key mediators of intercellular communication, have been increasingly recognized for their role in the oncogenic processes, particularly in facilitating drug resistance. This article delves into the emerging evidence linking exosomal lncRNAs to the modulation of drug resistance mechanisms in cancers such as ovarian, cervical, and endometrial cancer. It synthesizes current research findings on how these lncRNAs influence cancer cell survival, tumor microenvironment, and chemotherapy efficacy. Additionally, the review highlights potential therapeutic strategies targeting exosomal lncRNAs, proposing a new frontier in overcoming drug resistance. By mapping the interface of exosomal lncRNAs and drug resistance, this article aims to provide a comprehensive understanding that could pave the way for innovative treatments and improved patient outcomes in female reproductive system cancers.
Collapse
Affiliation(s)
- Nooshafarin Shirani
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Neda Abdi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Hajar Yaghoobi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
10
|
Kasiński D, Szeliski K, Drewa T, Pokrywczyńska M. Extracellular vesicles-a new player in the development of urinary bladder cancer. Ther Adv Med Oncol 2025; 17:17588359241297529. [PMID: 39850919 PMCID: PMC11755519 DOI: 10.1177/17588359241297529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/18/2024] [Indexed: 01/25/2025] Open
Abstract
Bladder cancer was the 10th most commonly diagnosed cancer worldwide in 2020. Extracellular vesicles (EVs) are nano-sized membranous structures secreted by all types of cells into the extracellular space. EVs can transport proteins, lipids, or nucleic acids to specific target cells. What brings more attention and potential implications is the fact that cancer cells secrete more EVs than non-malignant cells. EVs are widely studied for their role in cancer development. This publication summarizes the impact of EVs secreted by urinary bladder cancer cells on urinary bladder cancer development and metastasis. EVs isolated from urinary bladder cancer cells affect other lower-grade cancer cells or normal cells by inducing different metabolic pathways (transforming growth factor β/Smads pathway; phosphoinositide 3-kinase/Akt pathway) that promote epithelial-mesenchymal transition. The cargo carried by EVs can also induce angiogenesis, another critical element in the development of bladder cancer, and modulate the immune system response in a tumor-beneficial manner. In summary, the transfer of substances produced by tumor cells via EVs to the environment influences many stages of tumor progression. An in-depth understanding of the role EVs play in the development of urinary bladder cancer is crucial for the development of future anticancer therapies.
Collapse
Affiliation(s)
- Damian Kasiński
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Jagiellońska 13/15, 85-067 Bydgoszcz, Poland
| | - Kamil Szeliski
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Drewa
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Marta Pokrywczyńska
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
11
|
ZHOU ZHENGYANG, QIAO LEI, WANG TONGTONG, PAN WEN, DUAN JINGJING, ZHANG HAIYANG, DENG TING, BA YI, HE YI. Exosomal miR-224-3p promotes lymphangiogenesis and lymph node metastasis by targeting GSK3B in gastric cancer. Oncol Res 2025; 33:327-345. [PMID: 39866224 PMCID: PMC11753999 DOI: 10.32604/or.2024.050431] [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: 02/02/2024] [Accepted: 05/27/2024] [Indexed: 01/28/2025] Open
Abstract
Background Patients with gastric cancer (GC) are prone to lymph node metastasis (LNM), which is an important factor for recurrence and poor prognosis of GC. Nowadays, more and more studies have confirmed that exosomes can participate in tumor lymphangiogenesis. An in-depth exploration of the pathological mechanism in the process of LNM in GC may provide effective targets and improve the diagnosis and treatment effect. Materials and Methods We used sequencing analysis of collected serum to screen out exo-miRNA related to LNM in GC. ELISA, qRT-PCR, Western Blot, RNA pull-down assay, Transwell assay, animal experiments, and other experiments were used to verify the results. Results In this study, we screened out miR-224-3p related to GC progression and LNM in a vascular endothelial growth Factor C (VEGFC)-independent manner. We found that exo-miR-224-3p derived from GC cells could enter human lymphatic endothelial cells (HLECs) and promote the tube formation and migration of HLECs. In addition, it was revealed that miR-224-3p could bind to the 3'UTR region of GSK3B mRNA. Then, we proved that inhibiting the expression of GSK3B could suppress the phosphorylation of β-catenin and promote the transcription of PROX1, thus leading to tumor lymphangiogenesis. Furthermore, it was also found that hnRNPA1 mediated the sorting of miR-224-3p into exosomes, and the high expression of PKM2 promoted the secretion of exo-miR-224-3p. Conclusions Our discovery of the exo-miR-224-3p/GSK3B/β-catenin/PROX1 axis may provide a new direction for the clinical treatment of GC.
Collapse
Affiliation(s)
- ZHENGYANG ZHOU
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - LEI QIAO
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - TONGTONG WANG
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - WEN PAN
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - JINGJING DUAN
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - HAIYANG ZHANG
- Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300060, China
| | - TING DENG
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - YI BA
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - YI HE
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| |
Collapse
|
12
|
Zang W, Yang Y, Chen J, Mao Q, Xue W, Hu Y. The MIR181A2HG/miR-5680/VCAN-CD44 Axis Regulates Gastric Cancer Lymph Node Metastasis by Promoting M2 Macrophage Polarization. Cancer Med 2025; 14:e70600. [PMID: 39823128 PMCID: PMC11739459 DOI: 10.1002/cam4.70600] [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: 02/16/2024] [Revised: 12/21/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Lymphatic metastasis in gastric cancer (GC) profoundly influences its prognosis, but the precise mechanism remains elusive. In this study, we identified the long noncoding RNA MIR181A2HG as being upregulated in GC and associated with LNs metastasis and prognosis. METHODS The expression of MIR181A2HG in GC was identified through bioinformatics screening analysis and qRT-PCR validation. Both in vitro and in vivo functional studies revealed that MIR181A2HG facilitates lymphangiogenesis and lymphatic metastasis. Techniques such as immunofluorescence, immunohistochemistry, qRT-PCR, ELISA, CHIP, RNA-pulldown, luciferase reporter assay, and Co-IP were employed to investigate the mechanism of MIR181A2HG in LNs metastasis of GC. RESULTS MIR181A2HG overexpressed in GC signifies an unfavorable prognosis and drives M2 polarization of TAMs enhancing lymphangiogenesis. Mechanistically, MIR181A2HG/miR-5680 axis as a novel ceRNA regulatory axis to upregulate versican (VCAN). On one hand, VCAN interacts with CD44 receptors on the surface of TAMs through paracrine secretion, promoting M2 macrophage polarization and subsequently enhancing the secretion of VEGF-C, ultimately facilitating lymphangiogenesis. On the other hand, VCAN binds to CD44 receptors on the surface of GC cells through autocrine secretion, activating the Hippo pathway and upregulating SP1, thereby promoting the transcription of MIR181A2HG and establishing a feedback loop driving lymphatic metastasis. CONCLUSION This study highlights the pivotal role of MIR181A2HG in GC progression and LNs metastasis. MIR181A2HG-based targeted therapy would represent a novel strategy for GC.
Collapse
Affiliation(s)
- Weijie Zang
- Department of Gastrointestinal SurgeryAffiliated Hospital and Medical School of Nantong UniversityNantongChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
- Nantong Key Laboratory of Gastrointestinal OncologyNantongChina
| | - Yongpu Yang
- Department of General SurgeryThe First Affiliated Hospital, Army Medical UniversityChongqingChina
- Department of Graduate SchoolDalian Medical UniversityDalianChina
| | - Junjie Chen
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
- Nantong Key Laboratory of Gastrointestinal OncologyNantongChina
| | - Qinsheng Mao
- Department of Gastrointestinal SurgeryAffiliated Hospital and Medical School of Nantong UniversityNantongChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
- Nantong Key Laboratory of Gastrointestinal OncologyNantongChina
| | - Wanjiang Xue
- Department of Gastrointestinal SurgeryAffiliated Hospital and Medical School of Nantong UniversityNantongChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
- Nantong Key Laboratory of Gastrointestinal OncologyNantongChina
| | - Yilin Hu
- Department of Gastrointestinal SurgeryAffiliated Hospital and Medical School of Nantong UniversityNantongChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
- Nantong Key Laboratory of Gastrointestinal OncologyNantongChina
| |
Collapse
|
13
|
Chen W, Mao Y, Zhan Y, Li W, Wu J, Mao X, Xu B, Shu F. Exosome-delivered NR2F1-AS1 and NR2F1 drive phenotypic transition from dormancy to proliferation in treatment-resistant prostate cancer via stabilizing hormonal receptors. J Nanobiotechnology 2024; 22:761. [PMID: 39695778 DOI: 10.1186/s12951-024-03025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer cells acquire the ability to reprogram their phenotype in response to targeted therapies, yet the transition from dormancy to proliferation in drug-resistant cancers remains poorly understood. In prostate cancer, we utilized high-plasticity mouse models and enzalutamide-resistant (ENZ-R) cellular models to elucidate NR2F1 as a key factor in lineage transition and ENZ resistance. Depletion of NR2F1 drives ENZ-R cells into a relative dormancy state, characterized by reduced proliferation and heightened drug resistance, while NR2F1 overexpression yields contrasting outcomes. Transcriptional sequencing analysis of NR2F1-silenced prostate cancer cells and tissues from the Cancer Genome Atlas-prostate cancer and SU2C cohorts indicated exosomes as the most enriched cell component, with pathways implicated in steroid hormone biosynthesis and drug metabolism. Moreover, NR2F1-AS1 forms a complex with SRSF1 to upregulate NR2F1 expression, facilitating its binding with ESR1 to sustain hormonal receptor expression and enhance proliferation in ENZ-R cells. Furthermore, HnRNPA2B1 interacts with NR2F1 and NR2F1-AS1, assisting their packaging into exosomes, wherein exosomal NR2F1 and NR2F1-AS1 promote the proliferation of dormant ENZ-R cells. Our works offer novel insights into the reawaking of dormant drug-resistant cancer cells governed by NR2F1 upregulation triggered by exosome-derived NR2F1-AS1 and NR2F1, suggesting therapeutic potential for phenotype reversal.
Collapse
Affiliation(s)
- Wenbin Chen
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yiyou Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - YiYuan Zhan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenfeng Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jun Wu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiangming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Fangpeng Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, National Children's Medical Center for South Central Region, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Wang Z, Li Z, Sun X, Men W, Xu Y. Cellular components of tumor microenvironment: understanding their role in lymphatic metastasis of tumors. Front Pharmacol 2024; 15:1463538. [PMID: 39726782 PMCID: PMC11670069 DOI: 10.3389/fphar.2024.1463538] [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: 07/12/2024] [Accepted: 10/28/2024] [Indexed: 12/28/2024] Open
Abstract
Metastasis is the leading cause of cancer-related death in cancer patients. Tumor cells primarily spread through the hematogenous and lymphatic system. The underlying mechanisms of hematogenous metastasis have been well described over the past few decades. However, the understanding of the molecular mechanisms involved in lymphatic metastasis is still at an early stage. Tumor microenvironment (TME), primarily consisting of T cells, B cells, tumor-associated macrophages, neutrophils, and cancer-associated fibroblasts, has been implicated in the development of lymphatic metastasis. Recent studies have been reported that the dynamic and complex interplay between these cellular components of TME has great effects on lymphatic metastasis. Here, we discussed the paradoxical roles of these cellular component within the TME during lymphatic metastasis, as well as potential therapeutic opportunities to re-educate these cells within the TME to have anti-tumorigenic effects.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zehui Li
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiangyu Sun
- Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Wanfu Men
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yan Xu
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
15
|
Lodha P, Acari A, Rieck J, Hofmann S, Dieterich LC. The Lymphatic Vascular System in Extracellular Vesicle-Mediated Tumor Progression. Cancers (Basel) 2024; 16:4039. [PMID: 39682225 DOI: 10.3390/cancers16234039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/28/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Tumor growth and progression require molecular interactions between malignant and host cells. In recent years, extracellular vesicles (EVs) emerged as an important pillar of such interactions, carrying molecular information from their donor cells to distant recipient cells. Thereby, the phenotype and function of the recipient cells are altered, which may facilitate tumor immune escape and tumor metastasis to other organs through the formation of pre-metastatic niches. A prerequisite for these effects of tumor cell-derived EVs is an efficient transport system from the site of origin to the body periphery. Here, we highlight the role of the lymphatic vascular system in the distribution and progression-promoting functions of tumor cell-derived EVs. Importantly, the lymphatic vascular system is the primary drainage system for interstitial fluid and its soluble, particulate, and cellular contents, and therefore represents the principal route for regional (i.e., to tumor-draining lymph nodes) and systemic distribution of EVs derived from solid tumors. Furthermore, recent studies highlighted the tumor-draining lymph node as a crucial site where tumor-derived EVs exert their effects. A deeper mechanistic understanding of how EVs gain access to the lymphatic vasculature, how they interact with their recipient cells in tumor-draining lymph nodes and beyond, and how they induce phenotypic and functional maladaptation will be instrumental to identify new molecular targets and conceive innovative approaches for cancer therapy.
Collapse
Affiliation(s)
- Pragati Lodha
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Heidelberg Bioscience International Graduate School (HBIGS), Faculty of Bioscience, Heidelberg University, 69120 Heidelberg, Germany
| | - Alperen Acari
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Heidelberg Bioscience International Graduate School (HBIGS), Faculty of Bioscience, Heidelberg University, 69120 Heidelberg, Germany
| | - Jochen Rieck
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Sarah Hofmann
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Lothar C Dieterich
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
16
|
Ma Y, Zhang X, Liu C, Zhao Y. Extracellular vesicles in cancers: mechanisms, biomarkers, and therapeutic strategies. MedComm (Beijing) 2024; 5:e70009. [PMID: 39611045 PMCID: PMC11604295 DOI: 10.1002/mco2.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 11/30/2024] Open
Abstract
Extracellular vesicles (EVs) composed of various biologically active constituents, such as proteins, nucleic acids, lipids, and metabolites, have emerged as a noteworthy mode of intercellular communication. There are several categories of EVs, including exosomes, microvesicles, and apoptotic bodies, which largely differ in their mechanisms of formation and secretion. The amount of evidence indicated that changes in the EV quantity and composition play a role in multiple aspects of cancer development, such as the transfer of oncogenic signals, angiogenesis, metabolism remodeling, and immunosuppressive effects. As EV isolation technology and characteristics recognition improve, EVs are becoming more commonly used in the early diagnosis and evaluation of treatment effectiveness for cancers. Actually, EVs have sparked clinical interest in their potential use as delivery vehicles or vaccines for innovative antitumor techniques. This review will focus on the function of biological molecules contained in EVs linked to cancer progression and their participation in the intricate interrelationship within the tumor microenvironment. Furthermore, the potential efficacy of an EV-based liquid biopsy and delivery cargo for treatment will be explored. Finally, we explicitly delineate the limitations of EV-based anticancer therapies and provide an overview of the clinical trials aimed at improving EV development.
Collapse
Affiliation(s)
- Yuxi Ma
- Cancer CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Precision Radiation OncologyWuhanChina
- Cancer CenterInstitute of Radiation OncologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaohui Zhang
- Cancer CenterHubei Key Laboratory of Cell HomeostasisCollege of Life SciencesTaiKang Center for Life and Medical SciencesWuhan UniversityWuhanChina
| | - Cuiwei Liu
- Cancer CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Precision Radiation OncologyWuhanChina
- Cancer CenterInstitute of Radiation OncologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yanxia Zhao
- Cancer CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Precision Radiation OncologyWuhanChina
- Cancer CenterInstitute of Radiation OncologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
17
|
Zhang D, Luo Y, Lin Y, Fang Z, Zheng H, An M, Xie Q, Wu Z, Yu C, Yang J, Yu M, Chen C, Chen R. Endosomal Trafficking Bypassed by the RAB5B-CD109 Interplay Promotes Axonogenesis in KRAS-Mutant Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405092. [PMID: 39488792 DOI: 10.1002/advs.202405092] [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: 05/10/2024] [Revised: 10/13/2024] [Indexed: 11/04/2024]
Abstract
Perineural invasion (PNI) represents a unique biological feature associated with poor prognosis in pancreatic ductal adenocarcinoma (PDAC), especially in the presence of KRAS mutations. Extracellular vesicle (EV)-packaged circular RNAs (circRNAs) function as essential mediators of tumor microenvironment communication, triggering PDAC cell invasion and distant metastasis. However, the regulatory mechanisms of EV-packaged circRNAs in the PNI of KRAS-mutant PDAC have not yet been elucidated. Herein, a KRASG12D mutation-responsive EV-packaged circRNA, circPNIT, which positively correlated with PNI in PDAC patients is identified. Functionally, KRASG12D PDAC-derived EV-packaged circPNIT promoted axonogenesis and PNI both in vitro and in vivo. Mechanistically, the circPNIT-mediated Rab5B-CD109 interplay bypassed traditional endosomal trafficking to anchor Rab5B to the lipid rafts of multivesicular bodies and packaged circPNIT into CD109+ EVs. Subsequently, CD109+ EVs delivered circPNIT to neurons by binding to TRPV1 and facilitating DSCAML1 transcription-induced axonogenesis, which in turn enhanced the PNI by activating the GFRα1/RET pathway. Importantly, circPNIT-loaded CD109+ EVs are established to dramatically promote PNI in a KRASG12D/+ Trp53R172H/+ Pdx-1-Cre mouse model. Collectively, the findings highlight the mechanism underlying how EV-packaged circRNAs mediate the PNI of KRAS-mutant PDAC cells through the Rab5B endosomal bypass, identifying circPNIT as an effective target for the treatment of neuro-metastatic PDAC.
Collapse
Affiliation(s)
- Dingwen Zhang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P. R. China
| | - Yuming Luo
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P. R. China
| | - Yan Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, 510120, P. R. China
| | - Zhou Fang
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P. R. China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510080, P. R. China
| | - Hanhao Zheng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, 510120, P. R. China
| | - Mingjie An
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, 510120, P. R. China
| | - Qingyu Xie
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P. R. China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510080, P. R. China
| | - Zhuo Wu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P. R. China
| | - Chao Yu
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P. R. China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510080, P. R. China
| | - Jiabin Yang
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P. R. China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510080, P. R. China
| | - Min Yu
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P. R. China
| | - Changhao Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, 510120, P. R. China
| | - Rufu Chen
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, P. R. China
| |
Collapse
|
18
|
Jaszek N, Bogdanowicz A, Siwiec J, Starownik R, Kwaśniewski W, Mlak R. Epigenetic Biomarkers as a New Diagnostic Tool in Bladder Cancer-From Early Detection to Prognosis. J Clin Med 2024; 13:7159. [PMID: 39685620 DOI: 10.3390/jcm13237159] [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: 10/22/2024] [Revised: 11/20/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Bladder cancer (BC) currently ranks as the 9th most common cancer worldwide. It is characterised by very high rates of recurrence and metastasis. Most cases of BC are of urothelial origin, and due to its ability to penetrate muscle tissue, BC is divided into non-muscle-invasive BC (NMIBC) and muscle-invasive BC (MIBC). The current diagnosis of BC is still based primarily on invasive cystoscopy, which is an expensive and invasive method that carries a risk of various complications. Urine sediment cytology is often used as a complementary test, the biggest drawback of which is its very low sensitivity concerning the detection of BC at early stages, which is crucial for prompt implementation of appropriate treatment. Therefore, there is a great need to develop innovative diagnostic techniques that would enable early detection and accurate prognosis of BC. Great potential in this regard is shown by epigenetic changes, which are often possible to observe long before the onset of clinical symptoms of the disease. In addition, these changes can be detected in readily available biological material, such as urine or blood, indicating the possibility of constructing non-invasive diagnostic tests. Over the past few years, many studies have emerged using epigenetic alterations as novel diagnostic and prognostic biomarkers of BC. This review provides an update on promising diagnostic biomarkers for the detection and prognosis of BC based on epigenetic changes such as DNA methylation and expression levels of selected non-coding RNAs (ncRNAs), taking into account the latest literature data.
Collapse
Affiliation(s)
- Natalia Jaszek
- Department of Laboratory Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Alicja Bogdanowicz
- Department of Laboratory Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Jan Siwiec
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Radosław Starownik
- Department of Urology and Urological Oncology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Wojciech Kwaśniewski
- Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Radosław Mlak
- Department of Laboratory Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
19
|
Ngowi EE, Lu T, Liu Q, Xie X, Wang N, Luo L, Deng L, Zhou Y, Zhang Z, Qiao A. Biofluid-Derived Exosomal LncRNAs: Their Potential in Obesity and Related Comorbidities. BIOLOGY 2024; 13:976. [PMID: 39765643 PMCID: PMC11673191 DOI: 10.3390/biology13120976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 01/03/2025]
Abstract
Obesity has escalated into a critical global health crisis, tripling in prevalence since the mid-1970s. This increase mirrors the rise in metabolic-associated diseases such as type 2 diabetes (T2D) and its complications, certain cancers, and cardiovascular conditions. While substantial research efforts have enriched our understanding and led to the development of innovative management strategies for these diseases, the suboptimal response rates of existing therapies remain a major obstacle to effectively managing obesity and its associated conditions. Over the years, inter-organ communication (IOC) has emerged as a crucial factor in the development and progression of metabolic disorders. Exosomes, which are nano-sized vesicular couriers released by cells, play a significant role in this communication by transporting proteins, lipids, and nucleic acids across cellular landscapes. The available evidence indicates that exosomal RNAs present in biofluids such as blood, urine, milk, vitreous humor (VH), and cerebrospinal fluid (CSF) are altered in numerous diseases, suggesting their diagnostic and therapeutic potential. Long non-coding RNAs contained in exosomes (exo-lncRNAs) have attracted considerable interest, owing to their ability to interact with critical components involved in a multitude of metabolic pathways. Recent studies have found that alterations in exo-lncRNAs in biofluids correlate with several metabolic parameters in patients with metabolic-associated conditions; however, their exact roles remain largely unclear. This review highlights the diagnostic and therapeutic potential of exosomal lncRNAs in obesity and its associated conditions, emphasizing their role in IOC and disease progression, aiming to pave the way for further research in this promising domain.
Collapse
Affiliation(s)
- Ebenezeri Erasto Ngowi
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Tuyan Lu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Qing Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Xianghong Xie
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Ning Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Liping Luo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Lijuan Deng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Yinghua Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Zhihong Zhang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Aijun Qiao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
20
|
Tang H, Liu X, Ke J, Tang Y, Luo S, Li XK, Huang M. New perspectives of exosomes in urologic malignancies - Mainly focus on biomarkers and tumor microenvironment. Pathol Res Pract 2024; 263:155645. [PMID: 39476607 DOI: 10.1016/j.prp.2024.155645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/09/2024] [Accepted: 10/02/2024] [Indexed: 11/10/2024]
Abstract
Bladder cancer (BCa) and renal cell carcinoma (RCC) are prevalent urologic malignancies (UM) characterized by high morbidity and frequent recurrence. Current diagnostic approaches, often invasive, often indicate an advanced disease stage. And the complex tumor microenvironment often promotes tumor progression and induces resistance to chemotherapy. Current diagnostic and therapeutic modalities often fail to achieve satisfactory outcomes for patients. Exosomes transport diverse cargoes, including cytokines, proteins, lipids, non-coding RNAs, and microRNAs, crucial for intercellular communication. Exosomes have shown potential as biomarkers for UM, participating in tumor progression, especially within the tumor microenvironment (TME), including tumor cell apoptosis, proliferation, migration, invasion, depletion of immune cell function, epithelial-mesenchymal transition (EMT), angiogenesis, and more.In this review, we summarize research advances related to exosomes in UM, focusing on the role of exosomes as biomarkers in bladder and renal cancer, highlighting their significance within the TME.
Collapse
Affiliation(s)
- Hai Tang
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xing Liu
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jingwei Ke
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yiquan Tang
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Songtao Luo
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xu Kun Li
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Mingwei Huang
- Urology department, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
21
|
Wang T, Zhang H. Exploring the roles and molecular mechanisms of RNA binding proteins in the sorting of noncoding RNAs into exosomes during tumor progression. J Adv Res 2024; 65:105-123. [PMID: 38030125 PMCID: PMC11518959 DOI: 10.1016/j.jare.2023.11.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/26/2023] [Accepted: 11/24/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND RNA binding proteins (RBPs) play a role in sorting non-coding RNAs (ncRNAs) into exosomes. These ncRNAs, carried by exosomes, are involved in regulating various aspects of tumor progression, including metastasis, angiogenesis, control of the tumor microenvironment, and drug resistance. Recent studies have emphasized the importance of the RBP-ncRNA-exosome mechanism in tumor regulation. AIM OF REVIEW This comprehensive review aims to explore the RBP-ncRNA-exosome mechanism and its influence on tumor development. By understanding this intricate mechanism provides novel insights into tumor regulation and may lead to innovative treatment strategies in the future. KEY SCIENTIFIC CONCEPTS OF REVIEW The review discusses the formation of exosomes and the complex relationships among RBPs, ncRNAs, and exosomes. The RBP-ncRNA-exosome mechanism is shown to affect various aspects of tumor biology, including metastasis, multidrug resistance, angiogenesis, the immunosuppressive microenvironment, and tumor progression. Tumor development relies on the transmission of information between cells, with RBPs selectively mediating sorting of ncRNAs into exosomes through various mechanisms, which in turn carry ncRNAs to regulate RBPs. The review also provides an overview of potential therapeutic strategies, such as targeted drug discovery and genetic engineering for modifying therapeutic exosomes, which hold great promise for improving cancer treatment.
Collapse
Affiliation(s)
- Ting Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
22
|
Lin Y, Zheng H, Jia L, Luo Y, Zhang D, An M, Pang M, Diao X, Li W, Chen J, Li Y, Liu D, Liu Z, Huang J, Lin T, Chen C. Integrin α6-containing extracellular vesicles promote lymphatic remodelling for pre-metastatic niche formation in lymph nodes via interplay with CD151. J Extracell Vesicles 2024; 13:e12518. [PMID: 39329462 PMCID: PMC11428163 DOI: 10.1002/jev2.12518] [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: 01/29/2024] [Revised: 08/08/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Heterogeneous extracellular vesicles (EVs) from various types of tumours are acknowledged for inducing the formation of pre-metastatic "niches" in draining lymph nodes (LNs) to promote lymphatic metastasis. In order to identify the specific subpopulations of EVs involved, we performed high-resolution proteomic analysis combined with nanoflow cytometry of bladder cancer (BCa) tissue-derived EVs to identify a novel subset of tumour-derived EVs that contain integrin α6 (ITGA6+EVs) and revealed the positive correlation of ITGA6+EVs with the formation of pre-metastatic niche in draining LNs and lymphatic metastasis in multicentre clinical analysis of 820-case BCa patients. BCa-derived ITGA6+EVs induced E-selectin (SELE)-marked lymphatic remodelling pre-metastatic niche and promoted metastasis in draining LNs through delivering cargo circRNA-LIPAR to lymphatic endothelial cells in vivo and in vitro. Mechanistically, LIPAR linked ITGA6 to the switch II domain of RAB5A and sustained RAB5A GTP-bound activated state, thus maintaining the production of ITGA6+EVs loaded with LIPAR through endosomal trafficking. ITGA6+EVs targeted lymphatic vessels through ITGA6-CD151 interplay and released LIPAR to induce SELE overexpression-marked lymphatic remodelling pre-metastatic niche. Importantly, we constructed engineered-ITGA6 EVs to inhibit lymphatic pre-metastatic niche, which suppressed lymphatic metastasis and prolonged survival in preclinical models. Collectively, our study uncovers the mechanism of BCa-derived ITGA6+EVs mediating pre-metastatic niche and provides an engineered-EV-based strategy against BCa lymphatic metastasis.
Collapse
Affiliation(s)
- Yan Lin
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Hanhao Zheng
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Linpei Jia
- Department of NephrologyXuanwu Hospital, Capital Medical UniversityBeijingP. R. China
| | - Yuming Luo
- Department of General SurgeryGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdongP. R. China
| | - Dingwen Zhang
- Department of General SurgeryGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdongP. R. China
| | - Mingjie An
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Mingrui Pang
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Xiayao Diao
- Department of Thoracic SurgeryPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wenjie Li
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Jiancheng Chen
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Yuanlong Li
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Daiyin Liu
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Zhicong Liu
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Jian Huang
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Tianxin Lin
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
- Department of UrologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdongP. R. China
| | - Changhao Chen
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial Hospital, State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| |
Collapse
|
23
|
Pedersen C, Chen VT, Herbst P, Zhang R, Elfert A, Krishan A, Azar DT, Chang JH, Hu WY, Kremsmayer TP, Jalilian E, Djalilian AR, Guaiquil VH, Rosenblatt MI. Target specification and therapeutic potential of extracellular vesicles for regulating corneal angiogenesis, lymphangiogenesis, and nerve repair. Ocul Surf 2024; 34:459-476. [PMID: 39426677 PMCID: PMC11921040 DOI: 10.1016/j.jtos.2024.10.005] [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: 08/01/2024] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Extracellular vesicles, including exosomes, are small extracellular vesicles that range in size from 30 nm to 10 μm in diameter and have specific membrane markers. They are naturally secreted and are present in various bodily fluids, including blood, urine, and saliva, and through the variety of their internal cargo, they contribute to both normal physiological and pathological processes. These processes include immune modulation, neuronal synapse formation, cell differentiation, cancer metastasis, angiogenesis, lymphangiogenesis, progression of infectious disease, and neurodegenerative disorders like Alzheimer's and Parkinson's disease. In recent years, interest has grown in the use of exosomes as a potential drug delivery system for various diseases and injuries. Importantly, exosomes originating from a patient's own cells exhibit minimal immunogenicity and possess remarkable stability along with inherent and adjustable targeting capabilities. This review explores the roles of exosomes in angiogenesis, lymphangiogenesis, and nerve repair with a specific emphasis on these processes within the cornea. Furthermore, it examines exosomes derived from specific cell types, discusses the advantages of exosome-based therapies in modulating these processes, and presents some of the most established methods for exosome isolation. Exosome-based treatments are emerging as potential minimally invasive and non-immunogenic therapies that modulate corneal angiogenesis and lymphangiogenesis, as well as enhance and accelerate endogenous corneal nerve repair.
Collapse
Affiliation(s)
- Cameron Pedersen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victoria T Chen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Paula Herbst
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Runze Zhang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Amr Elfert
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Abhi Krishan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| | - Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, IL, USA
| | - Tobias P Kremsmayer
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Elmira Jalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victor H Guaiquil
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
24
|
Farahani M, Ghazimoradi MH. Dissecting the roles of exosomal cancer-associated fibroblasts-derived non-coding RNAs in tumor progression: A complete guide. Pathol Res Pract 2024; 262:155576. [PMID: 39232286 DOI: 10.1016/j.prp.2024.155576] [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: 05/09/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Cancer-associated fibroblasts are the most important cellular component of the tumor microenvironment, controlling cancer progression and therapeutic response. These cells in the tumor microenvironment regulate tumor progression and development as oncogenic or tumor suppressor agents. However, the mechanisms by which CAFs communicate with cancer cells remain to investigate. Here, we review evidence that extracellular vesicles, particularly exosomes, serve as vehicles for the intercellular transfer of bioactive cargos, notably microRNAs and long non-coding RNAs, from CAFs to cancer cells. We try to highlight molecular pathways of non-coding RNAs and the interaction among these molecules. Together, these findings elucidate a critical exosome-based communication axis by which CAFs create mostly a supportive pro-tumorigenic microenvironment and highlight therapeutic opportunities for disrupting this intercellular crosstalk.
Collapse
Affiliation(s)
- Mahnaz Farahani
- Department of Neurology, School of Medicine, Hazrat Rasool-E Akram, General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad H Ghazimoradi
- Department of Molecular Genetics, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
25
|
Zhou X, Tong Y, Yu C, Pu J, Zhu W, Zhou Y, Wang Y, Xiong Y, Sun X. FAP positive cancer-associated fibroblasts promote tumor progression and radioresistance in esophageal squamous cell carcinoma by transferring exosomal lncRNA AFAP1-AS1. Mol Carcinog 2024; 63:1922-1937. [PMID: 38934786 DOI: 10.1002/mc.23782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are abundant and heterogeneous stromal cells in the tumor microenvironment, which play important roles in regulating tumor progression and therapy resistance by transferring exosomes to cancer cells. However, how CAFs modulate esophageal squamous cell carcinoma (ESCC) progression and radioresistance remains incompletely understood. The expression of fibroblast activation protein (FAP) in CAFs was evaluated by immunohistochemistry in 174 ESCC patients who underwent surgery and 78 pretreatment biopsy specimens of ESCC patients who underwent definitive chemoradiotherapy. We sorted CAFs according to FAP expression, and the conditioned medium (CM) was collected to culture ESCC cells. The expression levels of several lncRNAs that were considered to regulate ESCC progression and/or radioresistance were measured in exosomes derived from FAP+ CAFs and FAP- CAFs. Subsequently, cell counting kit-8, 5-ethynyl-2'-deoxyuridine, transwell, colony formation, and xenograft assays were performed to investigate the functional differences between FAP+ CAFs and FAP- CAFs. Finally, a series of in vitro and in vivo assays were used to evaluate the effect of AFAP1-AS1 on radiosensitivity of ESCC cells. FAP expression in stromal CAFs was positively correlated with nerve invasion, vascular invasion, depth of invasion, lymph node metastasis, lack of clinical complete response and poor survival. Culture of ESCC cells with CM/FAP+ CAFs significantly increased cancer proliferation, migration, invasion and radioresistance, compared with culture with CM/FAP- CAFs. Importantly, FAP+ CAFs exert their roles by directly transferring the functional lncRNA AFAP1-AS1 to ESCC cells via exosomes. Functional studies showed that AFAP1-AS1 promoted radioresistance by enhancing DNA damage repair in ESCC cells. Clinically, high levels of plasma AFAP1-AS1 correlated with poor responses to dCRT in ESCC patients. Our findings demonstrated that FAP+ CAFs promoted radioresistance in ESCC cells through transferring exosomal lncRNA AFAP1-AS1; and may be a potential therapeutic target for ESCC treatment.
Collapse
Affiliation(s)
- Xilei Zhou
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yusuo Tong
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Changhua Yu
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Juan Pu
- Department of Radiation Oncology, Lianshui County People's Hospital, Kangda College of Nanjing Medical University, Huai'an, China
| | - Weiguo Zhu
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yun Zhou
- Department of Radiotherapy, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, China
| | - Yuandong Wang
- Department of Radiotherapy, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Yaozu Xiong
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Zang Y, Li J, Wan B, Tai Y, Liu H, Li Q, Ji Y, Wang G. LOC730101 transmitted by exosomes facilitates laryngeal squamous cell carcinoma tumorigenesis via regulation of p38 MAPK gamma. Cell Signal 2024; 122:111336. [PMID: 39121975 DOI: 10.1016/j.cellsig.2024.111336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a prevalent human cancer with a complex pathogenesis that remains incompletely understood. Here, we unveil a long non-coding RNA (lncRNA) associated with LSCC tumorigenesis and progression. LOC730101 exhibits significant overexpression in human LSCC tissues, and elevated LOC730101 levels correlate with malignant clinicopathological characteristics. Moreover, we demonstrate that LOC730101 is encapsulated into exosomes in an hnRNPA2B1-dependent manner, serving as a promising plasma biomarker for discriminating LSCC patients from healthy individuals (AUC = 0.92 with 89.36% sensitivity and 86.36% specificity). Exosomes derived from LSCC cells enhance the viability, DNA synthesis rate, and invasiveness of normal nasopharynx epithelial cells, with pronounced effects observed upon LOC730101 overexpression. Additionally, exosomal LOC730101 promotes tumor growth in vivo. Mechanistically, exosomal LOC730101 internalization by normal nasopharynx epithelial cells leads to increased H3K4me3 levels on the p38 MAPK gamma (p38γ) promoter via direct interaction with hnRNPA2B1. This interaction activates p38γ transcription, ultimately driving LSCC tumorigenesis. Collectively, our findings uncover a novel exosomal lncRNA that mediates communication between normal and LSCC cells during LSCC carcinogenesis, suggesting that targeting LOC730101 may represent a promising therapeutic strategy for LSCC treatment.
Collapse
Affiliation(s)
- Yanzi Zang
- Department of Otolaryngology, People's Hospital of Henan Province, Zhengzhou City, 450003, China
| | - Jing Li
- Department of Otolaryngology, People's Hospital of Henan Province, Zhengzhou City, 450003, China
| | - Baoluo Wan
- Department of Otolaryngology, People's Hospital of Henan Province, Zhengzhou City, 450003, China
| | - Yong Tai
- Department of Otolaryngology, People's Hospital of Henan Province, Zhengzhou City, 450003, China
| | - Hongjian Liu
- Department of Otolaryngology, People's Hospital of Henan Province, Zhengzhou City, 450003, China
| | - Qian Li
- Department of Otolaryngology, People's Hospital of Henan Province, Zhengzhou City, 450003, China
| | - Yuzi Ji
- Department of Otolaryngology, People's Hospital of Henan Province, Zhengzhou City, 450003, China
| | - Guangke Wang
- Department of Otolaryngology, People's Hospital of Henan Province, Zhengzhou City, 450003, China.
| |
Collapse
|
27
|
Zhou Y, Zhao K, Li J, Peng C, Jin J, Chen J, Li Y, Xu G, Pan S. LINC00461 promotes bladder cancer cells EMT through miR-518b/HNRNPUL1 axis. Discov Oncol 2024; 15:419. [PMID: 39254804 PMCID: PMC11387575 DOI: 10.1007/s12672-024-01294-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Bladder cancer (BC) is a prevalent type of tumor in the urinary system, and it has been discovered that long non-coding RNA (lncRNA) plays a significant role in its occurrence and development. However, thus far, no reports have been published on the involvement of LINC00461 in BC. Here, we found that LINC00461 levels were upregulated in BC tissues and cell lines. Besides, knockdown of LINC00461 inhibited BC cell proliferation, migration, invasion through epithelial-mesenchymal transition (EMT), and slowed down tumor growth in vivo. Moreover, we found that LINC00461 regulated HNRNPUL1 expression through miR-518b sponge activity, and the miR-518 inhibitor could reverse the inhibitory effects of LINC00461 knockdown on BC cell proliferation, migration, and EMT. Our results suggest that LINC00461 may serve as a potential biomarker and therapeutic target for BC.
Collapse
Affiliation(s)
- Yijie Zhou
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Keyuan Zhao
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Junlong Li
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Chao Peng
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Jing Jin
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Jiajun Chen
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Yulei Li
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Gang Xu
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China.
| | - Shouhua Pan
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
28
|
García-Silva S, Peinado H. Mechanisms of lymph node metastasis: An extracellular vesicle perspective. Eur J Cell Biol 2024; 103:151447. [PMID: 39116620 DOI: 10.1016/j.ejcb.2024.151447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/12/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
In several solid tumors such as breast cancer, prostate cancer, colorectal cancer or melanoma, tumor draining lymph nodes are the earliest tissues where colonization by tumor cells is detected. Lymph nodes act as sentinels of metastatic dissemination, the deadliest phase of tumor progression. Besides hematogenous dissemination, lymphatic spread of tumor cells has been demonstrated, adding more complexity to the mechanisms involved in metastasis. A network of blood and lymphatic vessels surrounds tumors providing routes for tumor soluble factors to mediate regional and long-distance effects. Additionally, extracellular vesicles (EVs), particularly small EVs/exosomes, have been shown to circulate through the blood and lymph, favoring the formation of pre-metastatic niches in the tumor-draining lymph nodes (TDLNs) and distant organs. In this review, we present an overview of the relevance of lymph node metastasis, the structural and immune changes occurring in TDLNs during tumor progression, and how extracellular vesicles contribute to modulating some of these alterations while promoting the formation of lymph node pre-metastatic niches.
Collapse
Affiliation(s)
- Susana García-Silva
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain.
| | - Héctor Peinado
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| |
Collapse
|
29
|
Zhou H, Hu S, Yan W. Extracellular vesicles as modifiers of epigenomic profiles. Trends Genet 2024; 40:797-809. [PMID: 38845265 DOI: 10.1016/j.tig.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 09/12/2024]
Abstract
Extracellular vesicles (EVs), emerging as novel mediators between intercellular communication, encapsulate distinct bioactive cargoes to modulate multiple biological events, such as epigenetic remodeling. In essence, EVs and epigenomic profiles are tightly linked and reciprocally regulated. Epigenetic factors, including histone and DNA modifications, noncoding RNAs, and protein post-translational modifications (PTMs) dynamically regulate EV biogenesis to contribute to EV heterogeneity. Alternatively, EVs actively modify DNA, RNA, and histone profiles in recipient cells by delivering RNA and protein cargoes for downstream epigenetic enzyme regulation. Moreover, EVs display great potential as diagnostic markers and drug-delivery vehicles for therapeutic applications. The combination of parental cell epigenomic modification with single EV characterization would be a promising strategy for EV engineering to enhance the epidrug loading efficacy and accuracy.
Collapse
Affiliation(s)
- Haifeng Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Sheng Hu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Wei Yan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China..
| |
Collapse
|
30
|
Liu X, Wu F, Pan W, Liu G, Zhang H, Yan D, Zheng S, Ma Z, Ren X. Tumor-associated exosomes in cancer progression and therapeutic targets. MedComm (Beijing) 2024; 5:e709. [PMID: 39247621 PMCID: PMC11380050 DOI: 10.1002/mco2.709] [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: 09/11/2023] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Exosomes are small membrane vesicles that are released by cells into the extracellular environment. Tumor-associated exosomes (TAEs) are extracellular vesicles that play a significant role in cancer progression by mediating intercellular communication and contributing to various hallmarks of cancer. These vesicles carry a cargo of proteins, lipids, nucleic acids, and other biomolecules that can be transferred to recipient cells, modifying their behavior and promoting tumor growth, angiogenesis, immune modulation, and drug resistance. Several potential therapeutic targets within the TAEs cargo have been identified, including oncogenic proteins, miRNAs, tumor-associated antigens, immune checkpoint proteins, drug resistance proteins, and tissue factor. In this review, we will systematically summarize the biogenesis, composition, and function of TAEs in cancer progression and highlight potential therapeutic targets. Considering the complexity of exosome-mediated signaling and the pleiotropic effects of exosome cargoes has challenge in developing effective therapeutic strategies. Further research is needed to fully understand the role of TAEs in cancer and to develop effective therapies that target them. In particular, the development of strategies to block TAEs release, target TAEs cargo, inhibit TAEs uptake, and modulate TAEs content could provide novel approaches to cancer treatment.
Collapse
Affiliation(s)
- Xiaomin Liu
- Lab for Noncoding RNA & Cancer School of Life Sciences Shanghai University Shanghai China
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Fan Wu
- Lab for Noncoding RNA & Cancer School of Life Sciences Shanghai University Shanghai China
| | - Wei Pan
- Lab for Noncoding RNA & Cancer School of Life Sciences Shanghai University Shanghai China
| | - Guangchao Liu
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Hui Zhang
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Dawei Yan
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Saijing Zheng
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Zhongliang Ma
- Lab for Noncoding RNA & Cancer School of Life Sciences Shanghai University Shanghai China
| | - Xiaojun Ren
- Department of Chemistry College of Chemistry and Life Sciences Beijing University of Technology Beijing China
| |
Collapse
|
31
|
Yoon JH, Byun HJ, Kim SY, Jung DH, Lee SK. Exosomal LINC00853 promotes progression of gastric cancer via the MAP17/PDZK1/AKT signaling pathway. Noncoding RNA Res 2024; 9:876-886. [PMID: 38586313 PMCID: PMC10997811 DOI: 10.1016/j.ncrna.2024.03.011] [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: 01/06/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024] Open
Abstract
Although rare, there is ongoing research into biomarkers that predict the onset and recurrence of gastric cancer, particularly focusing on substances found in exosomes. Long non-coding RNAs (lncRNAs) have garnered attention for their potential in diagnosing gastric cancer. This study investigates the role of lncRNAs in gastric cancer, focusing on their presence in exosomes as potential biomarkers for the disease's onset and recurrence. We utilized the ArrayStar Human LncRNA array 2.0 to analyze lncRNA expression in tissues from early-stage gastric cancer patients. Our analysis highlighted LINC00853, which was significantly upregulated in cancer tissues and implicated in promoting epithelial-mesenchymal transition via the MAP17/PDZK1/AKT pathway. Functional studies on AGS and MKN74 gastric cancer cell lines demonstrated that LINC00853 facilitates cell proliferation, invasion, and migration. Additionally, RNA immunoprecipitation and electrophoretic mobility shift assays confirmed LINC00853 interaction with MAP17. Importantly, LINC00853 was also detected in exosomes from both patient samples and cell lines, and its downregulation led to decreased tumorigenicity in AGS cells. These findings suggest that both cellular and exosomal LINC00853 contribute to gastric cancer pathogenesis and may serve as valuable biomarkers for the disease.
Collapse
Affiliation(s)
| | | | - Seo Yeon Kim
- Department of Internal Medicine, Yonsei Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Da Hyun Jung
- Department of Internal Medicine, Yonsei Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Kil Lee
- Department of Internal Medicine, Yonsei Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
32
|
Zhao J, Ma Y, Zheng X, Sun Z, Lin H, Du C, Cao J. Bladder cancer: non-coding RNAs and exosomal non-coding RNAs. Funct Integr Genomics 2024; 24:147. [PMID: 39217254 DOI: 10.1007/s10142-024-01433-9] [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: 07/28/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Bladder cancer (BCa) is a highly prevalent type of cancer worldwide, and it is responsible for numerous deaths and cases of disease. Due to the diverse nature of this disease, it is necessary to conduct significant research that delves deeper into the molecular aspects, to potentially discover novel diagnostic and therapeutic approaches. Lately, there has been a significant increase in the focus on non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), due to their growing recognition for their involvement in the progression and manifestation of BCa. The interest in exosomes has greatly grown due to their potential for transporting a diverse array of active substances, including proteins, nucleic acids, carbohydrates, and lipids. The combination of these components differs based on the specific cell and its condition. Research indicates that using exosomes could have considerable advantages in identifying and forecasting BCa, offering a less invasive alternative. The distinctive arrangement of the lipid bilayer membrane found in exosomes is what makes them particularly effective for administering treatments aimed at managing cancer. In this review, we have tried to summarize different ncRNAs that are involved in BCa pathogenesis. Moreover, we highlighted the role of exosomal ncRNAs in BCa.
Collapse
Affiliation(s)
- Jingang Zhao
- Department of Urology, Hangzhou Mingzhou Hospital, Hangzhou, 311215, Zhe'jiang, China
| | - Yangyang Ma
- Department of Urology, Hangzhou Mingzhou Hospital, Hangzhou, 311215, Zhe'jiang, China
| | - Xiaodong Zheng
- Department of the First Surgery, Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Force, Hangzhou, 310051, Zhe'jiang, China
| | - Zhen Sun
- Department of the First Surgery, Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Force, Hangzhou, 310051, Zhe'jiang, China
| | - Hongxiang Lin
- Department of Urology, Ganzhou Donghe Hospital, Ganzhou, 341000, Jiang'xi, China
| | - Chuanjun Du
- Department of Urology, Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310009, Zhe'jiang, China
| | - Jing Cao
- Department of Urology, Hangzhou Mingzhou Hospital, Hangzhou, 311215, Zhe'jiang, China.
| |
Collapse
|
33
|
Din MAU, Wan A, Chu Y, Zhou J, Yan Y, Xu Z. Therapeutic role of extracellular vesicles from human umbilical cord mesenchymal stem cells and their wide therapeutic implications in inflammatory bowel disease and other inflammatory disorder. Front Med (Lausanne) 2024; 11:1406547. [PMID: 39139783 PMCID: PMC11319305 DOI: 10.3389/fmed.2024.1406547] [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: 03/25/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
The chronic immune-mediated inflammatory condition known as inflammatory bowel disease (IBD) significantly affects the gastrointestinal system. While the precise etiology of IBD remains elusive, extensive research suggests that a range of pathophysiological pathways and immunopathological mechanisms may significantly contribute as potential factors. Mesenchymal stem cells (MSCs) have shown significant potential in the development of novel therapeutic approaches for various medical conditions. However, some MSCs have been found to exhibit tumorigenic characteristics, which limit their potential for medical treatments. The extracellular vesicles (EVs), paracrine factors play a crucial role in the therapeutic benefits conferred by MSCs. The EVs consist of proteins, microRNAs, and lipids, and are instrumental in facilitating intercellular communication. Due to the ease of maintenance, and decreased immunogenicity, tumorigenicity the EVs have become a new and exciting option for whole cell treatment. This review comprehensively assesses recent preclinical research on human umbilical cord mesenchymal stem cell (hUC-MSC)-derived EVs as a potential IBD therapy. It comprehensively addresses key aspects of various conditions, including diabetes, cancer, dermal injuries, neurological disorders, cardiovascular issues, liver and kidney diseases, and bone-related afflictions.
Collapse
Affiliation(s)
- Muhammad Azhar Ud Din
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine Jiangsu University, Zhenjiang, China
| | | | - Ying Chu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Jing Zhou
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Yongmin Yan
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Zhiliang Xu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| |
Collapse
|
34
|
Qin C, Li T, Lin C, Zhao B, Li Z, Zhao Y, Wang W. The systematic role of pancreatic cancer exosomes: distant communication, liquid biopsy and future therapy. Cancer Cell Int 2024; 24:264. [PMID: 39054529 PMCID: PMC11271018 DOI: 10.1186/s12935-024-03456-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
Pancreatic cancer remains one of the most lethal diseases worldwide. Cancer-derived exosomes, benefiting from the protective role of the lipid membrane, exhibit remarkable stability in the circulatory system. These exosomes, released by tumor microenvironment, contain various biomolecules such as proteins, RNAs, and lipids that plays a pivotal role in mediating distant communication between the local pancreatic tumor and other organs or tissues. They facilitate the transfer of oncogenic factors to distant sites, contributing to the compromised body immune system, distant metastasis, diabetes, cachexia, and promoting a microenvironment conducive to tumor growth and metastasis in pancreatic cancer patients. Beyond their intrinsic roles, circulating exosomes in peripheral blood can be detected to facilitate accurate liquid biopsy. This approach offers a novel and promising method for the diagnosis and management of pancreatic cancer. Consequently, circulating exosomes are not only crucial mediators of systemic cell-cell communication during pancreatic cancer progression but also hold great potential as precise tools for pancreatic cancer management and treatment. Exosome-based liquid biopsy and therapy represent promising advancements in the diagnosis and treatment of pancreatic cancer. Exosomes can serve as drug delivery vehicles, enhancing the targeting and efficacy of anticancer treatments, modulating the immune system, and facilitating gene editing to suppress tumor growth. Ongoing research focuses on biomarker identification, drug delivery systems, and clinical trials to validate the safety and efficacy of exosome-based therapies, offering new possibilities for early diagnosis and precision treatment in pancreatic cancer. Leveraging the therapeutic potential of exosomes, including their ability to deliver targeted drugs and modulate immune responses, opens new avenues for innovative treatment strategies.
Collapse
Affiliation(s)
- Cheng Qin
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianyu Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Lin
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bangbo Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zeru Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yutong Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weibin Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
35
|
Zhou J, Li L, Han Y, Ge G, Ji Q, Li H. RNA binding protein RALY facilitates colorectal cancer metastasis via enhancing exosome biogenesis in m6A dependent manner. Int J Biol Macromol 2024; 273:133112. [PMID: 38880454 DOI: 10.1016/j.ijbiomac.2024.133112] [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: 05/07/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Tumor metastasis is the leading cause of cancer-related death in patients with colorectal cancer (CRC). Heterogeneous nuclear ribonucleoproteins (hnRNPs) are RNA-binding proteins, involved in the tumorigenesis and metastasis of various cancers. However, the molecular mechanisms of hnRNPs in CRC metastasis remain unclear. This study aims to uncover the pivotal roles and molecular mechanisms of hnRNPs in CRC metastasis. Clinical database analysis suggested that the expression of hnRNP-Associated with Lethal Yellow (RALY, an important member of hnRNPs) was strongly correlated with the aggressiveness and survival of CRC patients. Gain- and loss-of-function studies demonstrated that RALY promotes the production of exosomes by increasing the formation of multivesicular bodies (MVBs) and enhancing the fusion of MVBs with the plasma membrane. Notably, RALY directly interacts with phospholipase D2 (PLD2) to enable exosome biogenesis, and cooperates with RBM15b to control PLD2 mRNA stability in an m6A-dependent manner. RALY-mediated exosome secretion activates pro-tumor macrophages and further facilitates CRC metastasis, while rescue experiments in vivo further confirmed that RALY-mediated exosome biogenesis facilitates CRC metastasis. Collectively, our findings demonstrate that RALY promotes exosome biogenesis and facilitates colorectal cancer metastasis by upregulating PLD2 and enhancing exosome production in an m6A-dependent manner, suggesting potential therapeutic strategies for combating CRC metastasis.
Collapse
Affiliation(s)
- Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ling Li
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yicun Han
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Qing Ji
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China.
| |
Collapse
|
36
|
Lu C, Gao H, Li H, Luo N, Fan S, Li X, Deng R, He D, Zhao H. A novel LINC02321 promotes cell proliferation and decreases cisplatin sensitivity in bladder cancer by regulating RUVBL2. Transl Oncol 2024; 45:101962. [PMID: 38677015 PMCID: PMC11066559 DOI: 10.1016/j.tranon.2024.101962] [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: 04/09/2023] [Revised: 10/15/2023] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
Bladder cancer (BC) has a high incidence and is prone to recurrence. In most instances, the low 5-year survival rate of advanced BC patients results from postoperative recurrence and drug resistance. Long noncoding RNAs (lncRNAs) can participate in numerous biological functions by regulating the expression of genes to affect tumorigenesis. Our previous work had demonstrated that a novel lncRNA, LINC02321, was associated with BC prognosis. In this study, A high expression of LINC02321 was found in BC tissues, which was associated with poor prognosis in patients. LINC02321 promoted both proliferation and G1-G0 progression in BC cells, while also inhibited sensitivity to cisplatin. Mechanistically, LINC02321 can bind to RUVBL2 and regulate the expression levels of RUVBL2 protein by affecting its half-life. RUVBL2 is involved in the DNA damage response. The potential of DNA damage repair pathways to exert chemosensitization has been demonstrated in vivo. The rescue experiment demonstrated that RUVBL2 overexpression can markedly abolish the decreased cell proliferation and the increased sensitivity of BC cells to cisplatin caused by LINC02321 knockdown. The results indicate that LINC02321 functions as an oncogene in BC, and may serve as a novel potential target for controlling BC progression and addressing cisplatin resistance in BC therapy.
Collapse
Affiliation(s)
- Chuncheng Lu
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming 650032, China
| | - Hongbin Gao
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming 650032, China
| | - Haiyuan Li
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming 650032, China
| | - Ning Luo
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming 650032, China
| | - Shipeng Fan
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming 650032, China
| | - Xi Li
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming 650032, China
| | - Renbin Deng
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming 650032, China
| | - Danpeng He
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming 650032, China
| | - Hui Zhao
- Department of Urology, The 1st Affiliated Hospital of Kunming Medical University, Kunming 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming 650032, China.
| |
Collapse
|
37
|
Liu J, Zhijin Z, Zhang W, Niraj M, Yang F, Changcheng G, Shen L, Xu T, Liu S, Junfeng Z, Mao S, Li W, Yao X. Urinary exosomes: Potential diagnostic markers and application in bladder cancer. Heliyon 2024; 10:e32621. [PMID: 38975179 PMCID: PMC11226776 DOI: 10.1016/j.heliyon.2024.e32621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
Background The exosome is a critical component of the intercellular communication., playing a vital role in regulating cell function. These small vesicles contain proteins, mRNAs, miRNAs, and lncRNAs, surrounded by lipid bilayer substances. Most cells in the human body can produce exosomes, released into various body fluids such as urine, blood, and cerebrospinal fluid. Bladder cancer is the most common tumor in the urinary system, with high recurrence and metastasis rates. Early diagnosis and treatment are crucial for improving patient outcomes. Methods This study employed the PubMed search engine to retrieve publicly accessible data pertaining to urinary exosomes. Results We summarize the origins and intricate biological characteristics of urinary exosomes, the introduction of research methodologies used in basic experiments to isolate and analyze these exosomes, the discussion of their applications and progress in the diagnosis and treatment of bladder cancer, and the exploration of the current limitations associated with using urinary exosomes as molecular biomarkers for diagnosing bladder cancer. Conclusion Exosomes isolated from urine may be used as molecular biomarkers for early detection of bladder cancer.
Collapse
Affiliation(s)
- Ji Liu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Zhang Zhijin
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Maskey Niraj
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Fuhan Yang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Guo Changcheng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Liliang Shen
- Department of Urology, The Affiliated Yinzhou Renmin Hospital of Medical School of Ningbo University, Ningbo, 315040, China
| | - Tianyuan Xu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Shenghua Liu
- Shanghai Huashan Hospital, Shanghai, 200433, China
| | - Zhang Junfeng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Shiyu Mao
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Wei Li
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, 200331, China
| |
Collapse
|
38
|
Wu H, Zeng C, Wu G, Fang F, Xiao C, Li L, Luo Y, Ouyang Z, Zhou C, Qian Y. Exosomal LRG1 promotes non-small cell lung cancer proliferation and metastasis by binding FN1 protein. Exp Cell Res 2024; 439:114097. [PMID: 38796135 DOI: 10.1016/j.yexcr.2024.114097] [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: 01/15/2024] [Revised: 05/13/2024] [Accepted: 05/19/2024] [Indexed: 05/28/2024]
Abstract
Leucine-rich α2-glycoprotein-1 (LRG1) is overexpressed in various cancers, including non-small cell lung cancer (NSCLC), but its role in NSCLC cell metastasis is not well understood. In this study, NSCLC cell exosomes were analyzed using different techniques, and the impact of exosomal LRG1 on NSCLC cell behavior was investigated through various assays both in vitro and in vivo. The study revealed that LRG1, found abundantly in NSCLC cells and exosomes, enhanced cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT). Exosomal LRG1 was shown to promote NSCLC cell metastasis in animal models. Additionally, the interaction between LRG1 and fibronectin 1 (FN1) in the cytoplasm was identified. It was observed that FN1 could counteract the effects of LRG1 knockdown on cell regulation induced by exosomes derived from NSCLC cells. Overall, the findings suggest that targeting exosomal LRG1 or FN1 may hold therapeutic potential for treating NSCLC.
Collapse
Affiliation(s)
- Hao Wu
- Department of Respiratory and Critical Care Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Chao Zeng
- Department of Thoracic Surgery, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Guodong Wu
- Department of Respiratory and Critical Care Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Fuyuan Fang
- Department of Respiratory and Critical Care Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Chunyang Xiao
- Department of Respiratory and Critical Care Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Liang Li
- Department of Respiratory and Critical Care Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Yisheng Luo
- Department of Respiratory and Critical Care Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Zezhong Ouyang
- Department of Respiratory and Critical Care Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Chaochao Zhou
- Department of Respiratory and Critical Care Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Youhui Qian
- Department of Respiratory and Critical Care Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| |
Collapse
|
39
|
Yang Z, Li X, Zhou L, Luo Y, Zhan N, Ye Y, Liu Z, Zhang X, Qiu T, Lin L, Peng L, Hu Y, Pan C, Sun M, Zhang Y. Ferroptosis-related lncRNAs: Distinguishing heterogeneity of the tumour microenvironment and predicting immunotherapy response in bladder cancer. Heliyon 2024; 10:e32018. [PMID: 38867969 PMCID: PMC11168393 DOI: 10.1016/j.heliyon.2024.e32018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024] Open
Abstract
Ferroptosis, a cell death pathway dependent on iron, has been shown in research to play a role in the development, advancement, and outlook of tumours through ferroptosis-related lncRNAs (FRLRs). However, the value of the FRLRs in bladder cancer (BLCA) has not been thoroughly investigated. This research project involved developing a predictive model using ten specific FRLRs (AC099850.4, AL731567.1, AL133415.1, AC021321.1, SPAG5-AS1, HMGA2-AS1, RBMS3-AS3, AC006160.1, AL583785.1, and AL662844.4) through univariate COX and LASSO regression techniques. The validation of this signature as a standalone predictor was confirmed in a group of 65 patients from the urology bladder tumour database at the First Affiliated Hospital of Wenzhou Medical University in Wenzhou, China. Patients were categorized based on their median risk score into either a low-risk group or a high-risk group. Enrichment analysis identified possible molecular mechanisms that could explain the variations in clinical outcomes observed in high-risk and low-risk groups. Moreover, we explored the correlation between FLPS and immunotherapy-related indicators. The ability of FLPS to forecast the effectiveness of immunotherapy was validated by the elevated levels of immune checkpoint genes (PD-L1, CTLA4, and PD-1) in the group at high risk. We also screened the crucial FRLR (HMGA2-AS1) through congruent expression and prognostic conditions and established a ceRNA network, indicating that HMGA2-AS1 may affect epithelial-mesenchymal transition by modulating the Wnt signalling pathway through the ceRNA mechanism. We identified the top five mRNAs (NFIB, NEGR1, JAZF1, JCAD, and ESM1) based on random forest algorithm and analysed the relationship between HMGA2-AS1, the top five mRNAs, and immunotherapy, and their interactions with drug sensitivities. Our results suggest that patients with BLCA have a greater sensitivity to four drugs (dasatinib, pazopanib, erismodegib and olaparib). Our study provides new insights into the TME, key signalling pathways, genome, and potential therapeutic targets of BLCA, with future guidance for immunotherapy and targeted precision drugs.
Collapse
Affiliation(s)
- Zhan Yang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaoqi Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lijun Zhou
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yaxian Luo
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Ning Zhan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yifan Ye
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhichao Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Xiaoting Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Tao Qiu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Lining Lin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Lianjie Peng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yiming Hu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Chaoran Pan
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Mouyuan Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yan Zhang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| |
Collapse
|
40
|
Phillips D, Noble D. Bubbling beyond the barrier: exosomal RNA as a vehicle for soma-germline communication. J Physiol 2024; 602:2547-2563. [PMID: 37936475 DOI: 10.1113/jp284420] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023] Open
Abstract
'Weismann's barrier' has restricted theories of heredity to the transmission of genomic variation for the better part of a century. However, the discovery and elucidation of epigenetic mechanisms of gene regulation such as DNA methylation and histone modifications has renewed interest in studies on the inheritance of acquired traits and given them mechanistic plausibility. Although it is now clear that these mechanisms allow many environmentally acquired traits to be transmitted to the offspring, how phenotypic information is communicated from the body to its gametes has remained a mystery. Here, we discuss recent evidence that such communication is mediated by somatic RNAs that travel inside extracellular vesicles to the gametes where they reprogram the offspring epigenome and phenotype. How gametes learn about bodily changes has implications not only for the clinic, but also for evolutionary theory by bringing together intra- and intergenerational mechanisms of phenotypic plasticity and adaptation.
Collapse
Affiliation(s)
- Daniel Phillips
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Denis Noble
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Xu G, Ban K, Mu H, Wang B. Human Umbilical Cord Mesenchymal Stem Cells-derived Exosomal lncRNA FAM99B Represses Hepatocellular Carcinoma Cell Malignancy. Mol Biotechnol 2024; 66:1389-1401. [PMID: 37351835 DOI: 10.1007/s12033-023-00795-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
Human umbilical cord mesenchymal stem cell (hucMSC)-derived exosomes (Exo) have been frequently investigated for disease control. This study was designed to explore the effects of hucMSC-Exo carrying lncRNA family with sequence similarity 99-member B (Exo-lncRNA FAM99B) on hepatocellular carcinoma (HCC) cell behaviour. The expression of lncRNA FAM99B in HCC cells was measured by reverse-transcription quantitative polymerase chain reaction. Protein levels of exosomal markers were quantified using western blotting. Flow cytometry analyses were performed to detect surface markers of hucMSCs and to measure the effects of Exo-lncRNA FAM99B on HCC cell cycle progression and cell apoptosis. Nanoparticle tracking analysis was used to measure the particle size of the exosomes. Additionally, cell viability was evaluated using methyl thiazolyl tetrazolium assays, and Transwell assays were performed to measure cell migration and invasion. Xenograft tumor models were established to explore the role of Exo-lncRNA FAM99B in vivo. Experimental results revealed that lncRNA FAM99B was downregulated in HCC cell lines, and low level of FAM99B is associated with poor survival rates in patients with HCC according to bioinformatics analysis. HucMSCs were identified in a good morphology with positively expressed CD105, CD29, and CD44 as well as negatively expressed CD31, CD14, and HLA-DR. High protein levels of exosomal markers (Alix, CD63 and TSG101) identified the existence of HucMSC-Exo. Importantly, the hucMSCs-Exo could enter HCC cells and exerted a suppressive effect on malignant cell activities. Moreover, overexpression of Exo-lncRNA FAM99B enhanced cell cycle arrest and cell apoptosis while suppressing cell viability, migration, and invasion in HCC. Exo-siRNA-FAM99B exerted the opposite effects on HCC cell process. In vivo experiments verified that Exo-lncRNA FAM99B inhibited tumorigenesis in HCC. In summary, lncRNA FAM99B derived from hucMSC-Exo inhibited malignant cellular phenotypes and tumorigenesis in HCC, which might provide a novel therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Gang Xu
- Department of General Surgery, Nanjing Tongren Hospital, No. 2007, Jiyin Avenue, Jiangning District, Nanjing, 211102, China
| | - Kunfeng Ban
- Department of General Surgery, Nanjing Tongren Hospital, No. 2007, Jiyin Avenue, Jiangning District, Nanjing, 211102, China
| | - Haifeng Mu
- Department of General Surgery, Nanjing Tongren Hospital, No. 2007, Jiyin Avenue, Jiangning District, Nanjing, 211102, China
| | - Baochen Wang
- Department of General Surgery, Nanjing Tongren Hospital, No. 2007, Jiyin Avenue, Jiangning District, Nanjing, 211102, China.
| |
Collapse
|
42
|
Arima J, Yoshino H, Fukumoto W, Kawahara I, Saito S, Li G, Fukuda I, Iizasa S, Mitsuke A, Sakaguchi T, Inoguchi S, Matsushita R, Nakagawa M, Tatarano S, Yamada Y, Enokida H. LncRNA BCYRN1 as a Potential Therapeutic Target and Diagnostic Marker in Serum Exosomes in Bladder Cancer. Int J Mol Sci 2024; 25:5955. [PMID: 38892143 PMCID: PMC11172611 DOI: 10.3390/ijms25115955] [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: 03/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Bladder cancer (BC) is a common genitourinary malignancy that exhibits silent morbidity and high mortality rates because of a lack of diagnostic markers and limited effective treatments. Here, we evaluated the role of the lncRNA brain cytoplasmic RNA 1 (BCYRN1) in BC. We performed loss-of-function assays to examine the effects of BCYRN1 downregulation in T24 and BOY BC cells. We found that BCYRN1 downregulation significantly inhibited the proliferation, migration, invasion, and three-dimensional spheroid formation ability and induced apoptosis in BC cells. Additionally, gene set enrichment analysis (GSEA) using RNA sequences from tumor fractions showed that BCYRN1 downregulation decreased the expression of mRNAs associated with the cell cycle. These findings were supported by observations of G2/M arrest in flow cytometry assays. Finally, we examined the expression of serum exosomal BCYRN1 as a biomarker. Clinically, BCYRN1 expression in serum exosomes from patients with BC (n = 31) was significantly higher than that in healthy donors (n = 19; mean difference: 4.1-fold higher, p < 0.01). Moreover, in patients who had undergone complete resection of BC, serum exosomal BCYRN1 levels were significantly decreased (n = 8). Thus, serum exosomal BCYRN1 may be a promising diagnostic marker and therapeutic target in patients with BC.
Collapse
Affiliation(s)
| | - Hirofumi Yoshino
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Li Y, Zhang S, Li Y, Liu J, Li Q, Zang W, Pan Y. The Regulatory Network of hnRNPs Underlying Regulating PKM Alternative Splicing in Tumor Progression. Biomolecules 2024; 14:566. [PMID: 38785973 PMCID: PMC11117501 DOI: 10.3390/biom14050566] [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: 03/30/2024] [Revised: 04/26/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
One of the hallmarks of cancer is metabolic reprogramming in tumor cells, and aerobic glycolysis is the primary mechanism by which glucose is quickly transformed into lactate. As one of the primary rate-limiting enzymes, pyruvate kinase (PK) M is engaged in the last phase of aerobic glycolysis. Alternative splicing is a crucial mechanism for protein diversity, and it promotes PKM precursor mRNA splicing to produce PKM2 dominance, resulting in low PKM1 expression. Specific splicing isoforms are produced in various tissues or illness situations, and the post-translational modifications are linked to numerous disorders, including cancers. hnRNPs are one of the main components of the splicing factor families. However, there have been no comprehensive studies on hnRNPs regulating PKM alternative splicing. Therefore, this review focuses on the regulatory network of hnRNPs on PKM pre-mRNA alternative splicing in tumors and clinical drug research. We elucidate the role of alternative splicing in tumor progression, prognosis, and the potential mechanism of abnormal RNA splicing. We also summarize the drug targets retarding tumorous splicing events, which may be critical to improving the specificity and effectiveness of current therapeutic interventions.
Collapse
Affiliation(s)
- Yuchao Li
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China; (Y.L.); (S.Z.); (J.L.); (Q.L.); (W.Z.)
| | - Shuwei Zhang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China; (Y.L.); (S.Z.); (J.L.); (Q.L.); (W.Z.)
| | - Yuexian Li
- Department of Radiation Oncology Gastrointestinal and Urinary and Musculoskeletal Cancer, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang 110042, China;
| | - Junchao Liu
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China; (Y.L.); (S.Z.); (J.L.); (Q.L.); (W.Z.)
| | - Qian Li
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China; (Y.L.); (S.Z.); (J.L.); (Q.L.); (W.Z.)
| | - Wenli Zang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China; (Y.L.); (S.Z.); (J.L.); (Q.L.); (W.Z.)
| | - Yaping Pan
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China; (Y.L.); (S.Z.); (J.L.); (Q.L.); (W.Z.)
| |
Collapse
|
44
|
Li H, Han X, Song L, Li X, Zhang L, Jin Z, Zhang Y, Wang T, Huang Z, Jia Z, Yang J. LINC00645 inhibits renal cell carcinoma progression by interacting with HNRNPA2B1 to regulate the ROCK1 mRNA stability. Gene 2024; 905:148232. [PMID: 38309317 DOI: 10.1016/j.gene.2024.148232] [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: 11/16/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
The lncRNA plays an important role in tumorigenesis and the progression of renal cell carcinoma (RCC). LINC00645 is one of the most different expressed lncRNA between RCC and normal renal tissue. However, the regulatory mechanism of LINC00645 in RCC remains unknown. Our results indicated that LINC00645 inhibited RCC proliferation, migration, and invasion. Mechanistically, HNRNPA2B1 directly bound to ROCK1 mRNA and strengthened its stability. LINC00645 competitively bound to the RRM1 domain, which is responsible for interacting with ROCK1 mRNA, reducing ROCK1 mRNA level by affecting posttranscriptional destabilization. The expression of LINC00645 was significantly reduced in RCC cells, significantly upregulating ROCK1 by abolishing the interaction with HNRNPA2B1, finally promoting RCC proliferation, migration, and invasion. Moreover, RCC cells with lower LINC00645 expression were more sensitive to the ROCK1 inhibitor Y-27632. Our study indicates that decreased expression of LINC00645 promotes the RCC progression via HNRNPA2B1/ROCK1 axis, providing a promising treatment strategy for RCC patients with decreased LINC00645 expression.
Collapse
Affiliation(s)
- Hao Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xu Han
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Liang Song
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiang Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Liwei Zhang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Zhibo Jin
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yu Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Tao Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Zhenlin Huang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Zhankui Jia
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Jinjian Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| |
Collapse
|
45
|
Liang H, Geng S, Wang Y, Fang Q, Xin Y, Li Y. Tumour-derived exosome SNHG17 induced by oestrogen contributes to ovarian cancer progression via the CCL13-CCR2-M2 macrophage axis. J Cell Mol Med 2024; 28:e18315. [PMID: 38680032 PMCID: PMC11056704 DOI: 10.1111/jcmm.18315] [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: 01/25/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024] Open
Abstract
Oestrogen is known to be strongly associated with ovarian cancer. There was much work to show the importance of lncRNA SNHG17 in ovarian cancer. However, no study has revealed the molecular regulatory mechanism and functional effects between oestrogen and SNHG17 in the development and metastasis of ovarian cancer. In this study, we found that SNHG17 expression was significantly increased in ovarian cancer and positively correlated with oestrogen treatment. Oestrogen could promote M2 macrophage polarization as well as ovarian cancer cells SKOV3 and ES2 cell exosomal SNHG17 expression. When exposure to oestrogen, exosomal SNHG17 promoted ovarian cancer cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) in vitro, and tumour growth and lung metastasis in vivo by accelerating M2-like phenotype of macrophages. Mechanically, exosomal SNHG17 could facilitate the release of CCL13 from M2 macrophage via the PI3K-Akt signalling pathway. Moreover, CCL13-CCR2 axis was identified to be involved in ovarian cancer tumour behaviours driven by oestrogen. There results demonstrate a novel mechanism that exosomal SNHG17 exerts an oncogenic effect on ovarian cancer via the CCL13-CCR2-M2 macrophage axis upon oestrogen treatment, of which SNHG17 may be a potential biomarker and therapeutic target for ovarian cancer responded to oestrogen.
Collapse
Affiliation(s)
- Haiyan Liang
- Department of Obstetrics and GynecologyChina‐Japan Friendship HospitalBeijingChina
| | - Shuo Geng
- Department of Obstetrics and GynecologyChina‐Japan Friendship HospitalBeijingChina
| | - Yadong Wang
- Scientific Research DepartmentGeneX Health Co., LtdBeijingChina
| | - Qing Fang
- Institute of Clinical MedicineChina‐Japan Friendship HospitalBeijingChina
| | - Yongfeng Xin
- Department of GynecologyThe People's Hospital of DaLaTeOrdosInner MongoliaChina
| | - Yanqing Li
- Department of GynecologyHebei Provincial Hospital of Traditional Chinese MedicineWuhanHebeiChina
| |
Collapse
|
46
|
Guo H, Fei L, Yu H, Li Y, Feng Y, Wu S, Wang Y. Exosome-encapsulated lncRNA HOTAIRM1 contributes to PM 2.5-aggravated COPD airway remodeling by enhancing myofibroblast differentiation. SCIENCE CHINA. LIFE SCIENCES 2024; 67:970-985. [PMID: 38332218 DOI: 10.1007/s11427-022-2392-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/20/2023] [Indexed: 02/10/2024]
Abstract
Emphysema, myofibroblast accumulation and airway remodeling can occur in the lungs due to exposure to atmospheric pollution, especially fine particulate matter (PM2.5), leading to chronic obstructive pulmonary disease (COPD). Specifically, bronchial epithelium-fibroblast communication participates in airway remodeling, which results in COPD. An increasing number of studies are now being conducted on the role of exosome-mediated cell-cell communication in disease pathogenesis. Here, we investigated whether exosomes generated from bronchial epithelial cells could deliver information to normal stromal fibroblasts and provoke cellular responses, resulting in airway obstruction in COPD. We studied the mechanism of exosome-mediated intercellular communication between human bronchial epithelial (HBE) cells and primary lung fibroblasts (pLFs). We found that PM2.5-induced HBE-derived exosomes promoted myofibroblast differentiation in pLFs. Then, the exosomal lncRNA expression profiles derived from PM2.5-treated HBE cells and nontreated HBE cells were investigated using an Agilent Human LncRNA Array. Combining coculture assays and direct exosome treatment, we found that HBE cell-derived exosomal HOTAIRM1 facilitated the myofibroblast differentiation of pLFs. Surprisingly, we discovered that exosomal HOTAIRM1 enhanced pLF proliferation to secrete excessive collagen secretion, leading to airway obstruction by stimulating the TGF-β/SMAD3 signaling pathway. Significantly, PM2.5 reduced FEV1/FVC and FEV1 and increased the level of serum exosomal HOTAIRM1 in healthy people; moreover, serum exosomal HOTAIRM1 was associated with PM2.5-related reductions in FEV1/FVC and FVC. These findings show that PM2.5 triggers alterations in exosome components and clarify that one of the paracrine mediators of myofibroblast differentiation is bronchial epithelial cell-derived HOTAIRM1, which has the potential to be an effective prevention and therapeutic target for PM2.5-induced COPD.
Collapse
Affiliation(s)
- Huaqi Guo
- The Ninth People's Hospital of Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Luo Fei
- The Ninth People's Hospital of Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Hengyi Yu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China
| | - Yan Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China
| | - Yan Feng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China
| | - Shaowei Wu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Xi'an Jiao Tong University Health Science Center, Xi'an, 710049, China.
| | - Yan Wang
- The Ninth People's Hospital of Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China.
| |
Collapse
|
47
|
Agnihotram R, Dhar R, Dhar D, Purushothaman K, Narasimhan AK, Devi A. Fusion of Exosomes and Nanotechnology: Cutting-Edge Cancer Theranostics. ACS APPLIED NANO MATERIALS 2024; 7:8489-8506. [DOI: 10.1021/acsanm.4c01033] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Rohan Agnihotram
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Debolina Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Kaavya Purushothaman
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Ashwin Kumar Narasimhan
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| |
Collapse
|
48
|
Yin C, Liufu C, Zhu T, Ye S, Jiang J, Wang M, Wang Y, Shi B. Bladder Cancer in Exosomal Perspective: Unraveling New Regulatory Mechanisms. Int J Nanomedicine 2024; 19:3677-3695. [PMID: 38681092 PMCID: PMC11048230 DOI: 10.2147/ijn.s458397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/06/2024] [Indexed: 05/01/2024] Open
Abstract
Bladder cancer, a prevalent malignant neoplasm of the urinary tract, exhibits escalating morbidity and mortality rates. Current diagnosis standards rely on invasive and costly cystoscopy and histopathology, underscoring the urgency for non-invasive, high-throughput, and cost-effective novel diagnostic techniques to ensure timely detection and standardized treatment. Recent years have witnessed the rise of exosome research in bladder cancer studies. Exosomes contain abundant bioactive molecules that can help elucidate the intricate mechanisms underlying bladder cancer pathogenesis and metastasis. Exosomes hold potential as biomarkers for early bladder cancer diagnosis while also serving as targeted drug delivery vehicles to enhance treatment efficacy and mitigate adverse effects. Furthermore, exosome analyses offer insights into the complex molecular signaling networks implicated in bladder cancer progression, revealing novel therapeutic targets. This review provides a comprehensive overview of prevalent exosome isolation techniques and highlights the promising clinical utility of exosomes in both diagnostic and therapeutic applications in bladder cancer management.
Collapse
Affiliation(s)
- Cong Yin
- Department of Urology, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
- Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Cen Liufu
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China
- Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Tao Zhu
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China
- Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Shuai Ye
- Department of Urology, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
- Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Jiahao Jiang
- Department of Urology, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
- Clinical College of Anhui Medical University, Shenzhen, People’s Republic of China
| | - Mingxia Wang
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China
| | - Yan Wang
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China
| | - Bentao Shi
- Department of Urology, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
| |
Collapse
|
49
|
Zhang Y, Zhang Y, Song J, Cheng X, Zhou C, Huang S, Zhao W, Zong Z, Yang L. Targeting the "tumor microenvironment": RNA-binding proteins in the spotlight in colorectal cancer therapy. Int Immunopharmacol 2024; 131:111876. [PMID: 38493688 DOI: 10.1016/j.intimp.2024.111876] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/04/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Colorectal cancer (CRC) is the third most common cancer and has the second highest mortality rate among cancers. The development of CRC involves both genetic and epigenetic abnormalities, and recent research has focused on exploring the ex-transcriptome, particularly post-transcriptional modifications. RNA-binding proteins (RBPs) are emerging epigenetic regulators that play crucial roles in post-transcriptional events. Dysregulation of RBPs can result in aberrant expression of downstream target genes, thereby affecting the progression of colorectal tumors and the prognosis of patients. Recent studies have shown that RBPs can influence CRC pathogenesis and progression by regulating various components of the tumor microenvironment (TME). Although previous research on RBPs has primarily focused on their direct regulation of colorectal tumor development, their involvement in the remodeling of the TME has not been systematically reported. This review aims to highlight the significant role of RBPs in the intricate interactions within the CRC tumor microenvironment, including tumor immune microenvironment, inflammatory microenvironment, extracellular matrix, tumor vasculature, and CRC cancer stem cells. We also highlight several compounds under investigation for RBP-TME-based treatment of CRC, including small molecule inhibitors such as antisense oligonucleotides (ASOs), siRNAs, agonists, gene manipulation, and tumor vaccines. The insights gained from this review may lead to the development of RBP-based targeted novel therapeutic strategies aimed at modulating the TME, potentially inhibiting the progression and metastasis of CRC.
Collapse
Affiliation(s)
- Yiwei Zhang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China; Queen Mary School, Nanchang University, 330006 Nanchang, China
| | - Yujun Zhang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China
| | - Jingjing Song
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China; School of Ophthalmology and Optometry of Nanchang University, China
| | - Xifu Cheng
- School of Ophthalmology and Optometry of Nanchang University, China
| | - Chulin Zhou
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Shuo Huang
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Wentao Zhao
- The 3rd Clinical Department of China Medical University, 10159 Shenyang, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China.
| | - Lingling Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China.
| |
Collapse
|
50
|
Tang L, Xu H, Wu T, Wu W, Lu Y, Gu J, Wang X, Zhou M, Chen Q, Sun X, Cai H. Advances in tumor microenvironment and underlying molecular mechanisms of bladder cancer: a systematic review. Discov Oncol 2024; 15:111. [PMID: 38602556 PMCID: PMC11009183 DOI: 10.1007/s12672-024-00902-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/21/2024] [Indexed: 04/12/2024] Open
Abstract
Bladder cancer is one of the most frequent malignant tumors of the urinary system. The prevalence of bladder cancer among men and women is roughly 5:2, and both its incidence and death have been rising steadily over the past few years. At the moment, metastasis and recurrence of advanced bladder cancer-which are believed to be connected to the malfunction of multigene and multilevel cell signaling network-remain the leading causes of bladder cancer-related death. The therapeutic treatment of bladder cancer will be greatly aided by the elucidation of these mechanisms. New concepts for the treatment of bladder cancer have been made possible by the advancement of research technologies and a number of new treatment options, including immunotherapy and targeted therapy. In this paper, we will extensively review the development of the tumor microenvironment and the possible molecular mechanisms of bladder cancer.
Collapse
Affiliation(s)
- Liu Tang
- Department of Nursing, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Haifei Xu
- Department of Urology, Nantong Tumor Hospital and Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Tong Wu
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China
| | - Wenhao Wu
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China
| | - Yuhao Lu
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China
| | - Jijia Gu
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China
| | - Xiaoling Wang
- Department of Urology, Nantong Tumor Hospital and Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Mei Zhou
- Department of Nursing, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China.
| | - Qiuyang Chen
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China.
| | - Xuan Sun
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China.
| | - Hongzhou Cai
- Department of Urology, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China.
| |
Collapse
|