1
|
Fujii N, Urabe F, Yamamoto S, Inoue K, Kimura T, Shiraishi K. Extracellular vesicles in renal cell carcinoma: A review of the current landscape and future directions. Urol Oncol 2025; 43:370-379. [PMID: 40069067 DOI: 10.1016/j.urolonc.2025.02.022] [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: 01/16/2025] [Accepted: 02/23/2025] [Indexed: 05/19/2025]
Abstract
Liquid biopsy, a minimally invasive biopsy method that uses patient body fluids (e.g., blood, urine, or saliva), is considered a useful biomarker for early diagnosis, monitoring of tumor progression, and evaluating treatment efficacy. Extracellular vesicles (EVs), a diverse group of particles classified according to their size and biosynthetic method, are liquid bilayer structures released from various cells. EVs contain specific information, such as DNA, RNA, and proteins derived from released cells. Consequently, they have attracted attention for use in liquid biopsy. EV-derived microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are useful biomarkers for cancer diagnosis, tumor progression, and drug treatment resistance. Renal cell carcinoma (RCC), one of the most common type of urological cancer, accounts for 90% of all renal tumors. In contrast to prostate cancer, for which a tumor marker has been established, clinically applicable and useful biomarkers remain to be established for RCC. EV-derived miRNAs and lncRNAs have been identified as useful biomarkers in several types of carcinoma for determining the diagnosis and predicting tumor progression, and drug treatment resistance in patients with RCC. The development and identification of biomarkers to diagnose and predict tumor progression in RCC will improve the management and prognosis of patients with RCC. This review focuses on EV-derived miRNAs and lncRNAs and discusses the currently available EV-based biomarkers in RCC and their future prospects.
Collapse
Affiliation(s)
- Nakanori Fujii
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan.
| | | | - Keiji Inoue
- Department of Urology, Kochi Medical School, Nankoku, Kochi, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Koji Shiraishi
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| |
Collapse
|
2
|
Saimaiti W, Ma J, Dilimulati P, Wang Y. Sunitinib-resistant renal cell carcinoma cell-derived exosomes promote facilitation of tumor progression via secretion of the lncRNA SNHG16. Hum Cell 2025; 38:100. [PMID: 40358820 DOI: 10.1007/s13577-025-01228-5] [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: 02/05/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
Renal cell carcinoma (RCC) is one of the most common tumors of high malignancy in the urological system. Sunitinib is commonly used to treat RCC, while drug resistance severely limited the therapeutic efficacy. Tumor-derived exosomes play important roles in facilitating cancer development. However, the role of drug-resistant tumor-derived exosomes in tumorigenesis and resistance of RCC has not been elucidated. Here we isolated sunitinib-sensitive/resistant RCC cells-derived exosomes, characterized by transmission electron microscopy (TEM) and western blot. Furthermore, co-culture experiments were performed and we found that sunitinib-resistant RCC cells-derived exosomes (R-exos) promoted cell proliferation and upregulated proliferation-related genes cyclin D1 (CCND1) and proliferating cell nuclear antigen (PCNA) expression, and inhibited apoptosis and the expression of Bax and Caspase-3 of sunitinib-resistant RCC (RCC/R) cells by delivering lncRNA small nuclear RNA host gene 16 (SNHG16). In resistant cell-derived xenograft (CDX-R) models, R-exos induced tumor growth in vivo, while knockdown of SNHG16 effectively diminished the tumorigenesis of RCC. Moreover, SNHG16 positively regulated the expression of trophinin associated protein (TROAP) by sponging miR-106a-5p in RCC cells, whereas inhibition of miR-106a-5p or overexpression of TROAP greatly reversed the suppression of tumorigenesis and sunitinib resistant by silencing SNHG16. R-exos lncRNA SNHG16 promoted sunitinib resistant and malignant progress by regulating the miR-106a-5p/TROAP axis, and targeting SNHG16/miR-106a-5p/TROAP axis may be a novel therapeutic approach for sunitinib-treated patients of RCC.
Collapse
Affiliation(s)
- WeiLijiang Saimaiti
- Department of Pediatric Urology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, 830054, China
| | - Jun Ma
- Department of Urology, First Affiliated Hospital, Xinjiang Medical University, No. 137, Liyuushan South Road, Urumqi, 830054, Xinjiang, China
| | - Paluoke Dilimulati
- Department of Pediatric Urology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, 830054, China
| | - Yujie Wang
- Department of Urology, First Affiliated Hospital, Xinjiang Medical University, No. 137, Liyuushan South Road, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
3
|
Bianchi N, Ancona P, Aguiari G. Molecular Mechanisms of Drug Resistance in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2025; 17:1613. [PMID: 40427113 PMCID: PMC12110057 DOI: 10.3390/cancers17101613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Renal cell carcinoma (RCC) accounts for about 3% of all human tumors. Alterations of oxygen, lipids, iron, and energy metabolism are involved in carcinogenesis, development, and expansion. Thirty percent of patients affected by clear cell renal cell carcinoma (ccRCC) will develop relapses or distance metastases (mRCC), dramatically reducing their life expectancy. Current first-line therapies for mRCC patients are based on treatment with immune checkpoint inhibitors (ICIs) alone and in combination with each other or with tyrosine kinase inhibitors (TKIs). However, only 20% of patients show a mild response because of innate or acquired drug resistance during long-term treatment; therefore, resistant patients need alternative first-line or second-line therapies. Pharmacological resistance represents a big problem that counteracts the efficacy of treatment by reducing overall survival (OS) in mRCC patients. Investigating the molecular mechanisms underlying drug resistance is crucial to overcoming drug insensitivity and enhancing therapeutic outcomes. In this review, we emphasize the latest and most significant studies on the molecular mechanisms that drive drug resistance in ccRCC carcinoma. Particular attention is given to the key signaling pathways involved in resistance, including those mediated by HIF, p53, Akt-mTOR, MEK-ERK cascades, Wnt signaling, autophagy, membrane transporters, ferroptosis, and non-coding RNAs. Understanding these resistance mechanisms is essential for developing new therapeutic strategies aimed to enhancing overall OS and improving the quality of life for mRCC patients. This review also discusses recent clinical trial findings on the use of specific inhibitors able to circumvent drug resistance. The data presented here could be valuable for clinicians in understanding the mechanisms of drug resistance, ultimately aiding in the management of ccRCC patients.
Collapse
Affiliation(s)
- Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (N.B.); (P.A.)
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (N.B.); (P.A.)
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
4
|
Liu G, Liu J, Li S, Zhang Y, He R. Exosome-Mediated Chemoresistance in Cancers: Mechanisms, Therapeutic Implications, and Future Directions. Biomolecules 2025; 15:685. [PMID: 40427578 PMCID: PMC12108986 DOI: 10.3390/biom15050685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Chemotherapy resistance represents a formidable obstacle in oncological therapeutics, substantially compromising the efficacy of adjuvant chemotherapy regimens and contributing to unfavorable clinical prognoses. Emerging evidence has elucidated the pivotal involvement of exosomes in the dissemination of chemoresistance phenotypes among tumor cells and within the tumor microenvironment. This review delineates two distinct intra-tumoral resistance mechanisms orchestrated by exosomes: (1) the exosome-mediated sequestration of chemotherapeutic agents coupled with enhanced drug efflux in neoplastic cells, and (2) the horizontal transfer of chemoresistance to drug-sensitive cells through the delivery of bioactive molecular cargo, thereby facilitating the propagation of resistance phenotypes across the tumor population. Furthermore, the review covers current in vivo experimental data focusing on targeted interventions against specific genetic elements and exosomal secretion pathways, demonstrating their potential in mitigating chemotherapy resistance. Additionally, the therapeutic potential of inhibiting exosome-mediated transporter transfer strategy is particularly examined as a promising strategy to overcome tumor resistance mechanisms.
Collapse
Affiliation(s)
| | | | | | - Yumiao Zhang
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| | - Ren He
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| |
Collapse
|
5
|
Lu X, Friedrich LJ, Efferth T. Natural products targeting tumour angiogenesis. Br J Pharmacol 2025; 182:2094-2136. [PMID: 37680009 DOI: 10.1111/bph.16232] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
Tumour angiogenesis is the formation of new blood vessels to support the growth of a tumour. This process is critical for tumour progression and metastasis, making it an attractive approach to cancer therapy. Natural products derived from plants, animals or microorganisms exert anti-angiogenic properties and can be used to inhibit tumour growth and progression. In this review, we comprehensively report on the current status of natural products against tumour angiogenesis from four perspectives until March 2023: (1) the role of pro-angiogenic factors and antiangiogenic factors in tumour angiogenesis; (2) the development of anti-tumour angiogenesis therapy (monoclonal antibodies, VEGFR-targeted small molecules and fusion proteins); (3) the summary of anti-angiogenic natural agents, including polyphenols, polysaccharides, alkaloids, terpenoids, saponins and their mechanisms of action, and (4) the future perspectives of anti-angiogenic natural products (bioavailability improvement, testing of dosage and side effects, combination use and discovery of unique natural-based compounds). Our review aims to better understand the potential of natural products for drug development in inhibiting tumour angiogenesis and further aid the effective transition of these outcomes into clinical trials. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lara Johanna Friedrich
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
6
|
Zheng M, Yang Z, Shi L, Zhao L, Liu K, Tang N. The role of lncRNAs in AKI and CKD: Molecular mechanisms, biomarkers, and potential therapeutic targets. Genes Dis 2025; 12:101509. [PMID: 40083322 PMCID: PMC11904545 DOI: 10.1016/j.gendis.2024.101509] [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: 04/03/2023] [Revised: 02/04/2024] [Accepted: 11/02/2024] [Indexed: 03/16/2025] Open
Abstract
Exosomes, a type of extracellular vesicle, are commonly found in different body fluids and are rich in nucleic acids (circRNA, lncRNAs, miRNAs, mRNAs, tRNAs, etc.), proteins, and lipids. They are involved in intercellular communication. lncRNAs are responsible for the modulation of gene expression, thus affecting the pathological process of kidney injury. This review summarizes the latest knowledge on the roles of exosome lncRNAs and circulating lncRNAs in the pathogenesis, biomarker discovery, and treatment of chronic kidney disease, renal fibrosis, and acute kidney injury, providing an overview of novel regulatory approaches and lncRNA delivery systems.
Collapse
Affiliation(s)
- Minhui Zheng
- Shanghai Innostar Bio-Technology Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Zixuan Yang
- Shanghai Innostar Bio-Technology Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Lei Shi
- Shanghai Innostar Bio-Technology Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Liyuan Zhao
- Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230000, China
- Yangtze Delta Drug Advanced Research Institute, Yangtze Delta Pharmaceutical College, Nantong, Jiangsu 226133, China
| | - Kelan Liu
- Intensive Care Unit, Liyang People's Hospital, Liyang, Jiangsu 213300, China
| | - Naping Tang
- Shanghai Innostar Bio-Technology Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| |
Collapse
|
7
|
Yin H, Shi J, Li S, You Q, Zhu H, Koo C, Liu B, Hou L, Wu C. Emerging roles of exosomal circRNAs in non-small cell lung cancer. J Transl Med 2025; 23:490. [PMID: 40307927 PMCID: PMC12042431 DOI: 10.1186/s12967-025-06463-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/06/2025] [Indexed: 05/02/2025] Open
Abstract
Despite the prevalence of non-small cell lung cancer (NSCLC) is high, the limited early detection and management of these tumors are restricted since there is an absence of reliable and precise diagnostic biomarkers and therapeutic targets. Exosomes transport functional molecules for facilitating intercellular communication, especially in the tumor microenvironment, indicating their potential as cancer biomarkers and therapeutic targets. Circular RNA (circRNA), a type of non-coding RNA possessing a covalently closed loop structure, substantial abundance, and tissue-specific expression patterns, is stably enriched in exosomes. In recent years, significant breakthroughs have been made in research on exosomal circRNA in NSCLC. This review briefly introduces the biogenesis, characterizations, and functions of circRNAs and exosomes, and systematically describes the biological functions and mechanisms of exosomal circRNAs in NSCLC. In addition, this study summarizes their role in the progression of NSCLC and discusses their clinical significance as biomarkers and therapeutic targets for NSCLC.
Collapse
Affiliation(s)
- Hongyuan Yin
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jiayi Shi
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shaoling Li
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qianhui You
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huici Zhu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chinying Koo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Baonian Liu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
8
|
Delshad M, Sanaei MJ, Mohammadi MH, Sadeghi A, Bashash D. Exosomal Biomarkers: A Comprehensive Overview of Diagnostic and Prognostic Applications in Malignant and Non-Malignant Disorders. Biomolecules 2025; 15:587. [PMID: 40305328 PMCID: PMC12024574 DOI: 10.3390/biom15040587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Exosomes are small extracellular vesicles, ranging from 30 to 150 nm, that are essential in cell biology, mediating intercellular communication and serving as biomarkers due to their origin from cells. Exosomes as biomarkers for diagnosing various illnesses have gained significant investigation due to the high cost and invasive nature of current diagnostic procedures. Exosomes have a clear advantage in the diagnosis of diseases because they include certain signals that are indicative of the genetic and proteomic profile of the ailment. This feature gives them the potential to be useful liquid biopsies for real-time, noninvasive monitoring, enabling early cancer identification for the creation of individualized treatment plans. According to our analysis, the trend toward utilizing exosomes as diagnostic and prognostic tools has raised since 2012. In this regard, the proportion of malignant indications is higher compared with non-malignant ones. To be precise, exosomes have been used the most in gastrointestinal, thoracic, and urogenital cancers, along with cardiovascular, diabetic, breathing, infectious, and brain disorders. To the best of our knowledge, this is the first research to examine all registered clinical trials that look at exosomes as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Mahda Delshad
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan 1411718541, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran;
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| |
Collapse
|
9
|
Li Z, Wang D, Zhu X. Roles of LncRNA ARSR in tumor proliferation, drug resistance, and lipid and cholesterol metabolism. Clin Transl Oncol 2025; 27:1356-1365. [PMID: 39251493 DOI: 10.1007/s12094-024-03700-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024]
Abstract
Cancer is one of the most serious diseases that threaten human life and health. Among all kinds of diseases, the mortality rate of malignant tumors is the second highest, second only to cardio-cerebrovascular diseases. Cancer treatment typically involves imaging, surgery, and pathological analysis. When patients are identified as carcinoma by the above means, there are often problems of distant metastasis, delayed treatment, and drug tolerance, indicating that patients have some poor prognosis and overall survival. Hence, the development of novel molecular biomarkers is of great clinical importance. In recent years, as an important mediator of material and information exchange between cells in the tumor microenvironment, lncRNA have attracted widespread attention for their roles in tumor development. In this review, we comprehensively summarize the up-to-date knowledge of lncARSR on diverse cancer types which mainly focuses on tumor proliferation, drug tolerance, and lipid and cholesterol metabolism, highlighting the potential of lncARSR as a diagnostic and prognostic biomarker and even a therapeutic target. In our final analysis, we provide a synthesized overview of the directions for future inquiry into lncARSR, and we are eager to witness the advancement of research that will elucidate the multifaceted nature of this lncRNA.
Collapse
Affiliation(s)
- Zhicheng Li
- Department of Urology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Dan Wang
- Department of Urology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Xiaojun Zhu
- Department of Urology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China.
| |
Collapse
|
10
|
Wei Z, Ye Y, Liu C, Wang Q, Zhang Y, Chen K, Cheng G, Zhang X. MIER2/PGC1A elicits sunitinib resistance via lipid metabolism in renal cell carcinoma. J Adv Res 2025; 70:287-305. [PMID: 38702028 PMCID: PMC11976417 DOI: 10.1016/j.jare.2024.04.032] [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: 02/04/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
INTRODUCTION Renal cell carcinoma (RCC) is one of the most common malignant tumors of the urinary system and accounts for more than 90 % of all renal tumors. Resistance to targeted therapy has emerged as a pivotal factor that contributes to the progressive deterioration of patients with advanced RCC. Metabolic reprogramming is a hallmark of tumorigenesis and progression, with an increasing body of evidence indicating that abnormal lipid metabolism plays a crucial role in the advancement of renal clear cell carcinoma. OBJECTIVES Clarify the precise mechanisms underlying abnormal lipid metabolism and drug resistance. METHODS Bioinformatics screening and analyses were performed to identify hub gene. qRT-PCR, western blot, chromatin immunoprecipitation (ChIP) assays, and other biological methods were used to explore and verify related pathways. Various cell line models and animal models were used to perform biological functional experiments. RESULTS In this study, we identified Mesoderm induction early response 2 (MIER2) as a novel biomarker for RCC, demonstrating its role in promoting malignancy and sunitinib resistance by influencing lipid metabolism in RCC. Mechanistically, MIER2 facilitated P53 deacetylation by binding to HDAC1. Acetylation modification augmented the DNA-binding stability and transcriptional function of P53, while deacetylation of P53 hindered the transcriptional process of PGC1A, leading to intracellular lipid accumulation in RCC. Furthermore, Trichostatin A (TSA), an inhibitor of HDAC1, was found to impede the MIER2/HDAC1/P53/PGC1A pathway, offering potential benefits for patients with sunitinib-resistant renal cell cancer. CONCLUSION Our findings highlight MIER2 as a key player in anchoring HDAC1 and inhibiting PGC1A expression through the deacetylation of P53, thereby inducing lipid accumulation in RCC and promoting drug resistance. Lipid-rich RCC cells compensate for energy production and sustain their own growth in a glycolysis-independent manner, evading the cytotoxic effects of targeted drugs and ultimately culminating in the development of drug resistance.
Collapse
Affiliation(s)
- Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhong Ye
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenchen Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunxuan Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailei Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Shenzhen Huazhong University of Science and Technology Research Institute, China.
| |
Collapse
|
11
|
Li X, Gong J, Ni X, Yin J, Zhang Y, Lv Z. Potential biological roles of exosomal non-coding RNAs in breast cancer. FASEB J 2025; 39:e70456. [PMID: 40079186 PMCID: PMC11904755 DOI: 10.1096/fj.202500022r] [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: 01/03/2025] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Breast cancer (BC) is one of the most common malignant tumors among women, accounting for 24.5% of all cancer cases and leading to 15.5% of cancer-related mortality. The treatment of BC patients remains a significant challenge due to the disease's high invasiveness, elevated metastatic potential, substantial drug resistance, and high recurrence rate. Exosomes, which are lipid-bilayer extracellular vesicles ranging in size from 30 to 150 nm, mediate intercellular communication between tumor cells and surrounding cells in the tumor microenvironment by transferring various bioactive substances, such as proteins, lipids, and nucleic acids. Recently, growing evidence has demonstrated that non-coding RNAs (ncRNAs) are enriched in exosomes and play a critical role in regulating cell proliferation, metastasis, drug resistance, and angiogenesis in BC. Consequently, exosomal ncRNAs have emerged as a promising therapeutic target for BC treatment, given their involvement in multiple processes of cancer progression. This review provides a comprehensive and in-depth analysis of emerging exosomal ncRNAs in BC, highlighting their potential biological mechanisms and advanced applications in BC treatment.
Collapse
Affiliation(s)
- Xiang Li
- Cancer CenterThe First Affiliated Hospital of Jilin UniversityChangchunJilinChina
| | - Junyi Gong
- Cancer CenterThe First Affiliated Hospital of Jilin UniversityChangchunJilinChina
| | - Xiang Ni
- Cancer CenterThe First Affiliated Hospital of Jilin UniversityChangchunJilinChina
| | - Junli Yin
- Cancer CenterThe First Affiliated Hospital of Jilin UniversityChangchunJilinChina
| | - Yi Zhang
- Cancer CenterThe First Affiliated Hospital of Jilin UniversityChangchunJilinChina
| | - Zheng Lv
- Cancer CenterThe First Affiliated Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
12
|
Zwamel AH, Ahmad AT, Altalbawy FMA, Malathi H, Singh A, Jabir MS, Aminov Z, Lal M, Kumar A, Jawad SF. Exosomal RNAs and EZH2: unraveling the molecular dialogue driving tumor progression. Med Oncol 2025; 42:103. [PMID: 40075013 DOI: 10.1007/s12032-025-02648-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
The EZH2 gene encodes an enzyme that is part of the epigenetic factor Polycomb Repressive Complex 2 (PRC2). In order to control gene expression, PRC2 mainly modifies chromatin structure. In this complex process, EZH2 methylates histone proteins, which in turn suppresses further RNA transcriptions. As a result, EZH2 dysregulations can occasionally induce abnormal gene expression patterns, which can aid in the development and progression of cancer. Non-coding RNAs significantly impact the expression of EZH2 through epigenetic mechanisms. Meanwhile, normal and cancerous cells frequently release vesicles into the extracellular matrix, also known as exosomes, that occasionally carry RNA molecules from their origin cells, including messenger RNAs, microRNAs, and other non-coding RNAs. Thus exosomes are granted the ability to regulate numerous physiological functions and act as crucial messengers between cells by influencing gene expression in the recipient cell. We conducted this review to focus on EZH2's substantial biological role and the mechanisms that regulate it, driven by the desire to understand the possible impact of exosomal RNAs on EZH2 expression.
Collapse
Affiliation(s)
- Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | | | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bengaluru, Karnataka, India
| | - Amandeep Singh
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, 140307, Punjab, India
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, Samarkand, Uzbekistan
| | - Madan Lal
- Department of Medicine, National Institute of Medical Sciences, NIMS University, Rajasthan, Jaipur, India
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg 620002, Russia
- Department of Technical Sciences, Western Caspian University, Baku, Azerbaijan
- Department of Mechanical Engineering, Karpagam Academy of Higher Education, Coimbatore, 641021, India
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, Babylon, Iraq
| |
Collapse
|
13
|
Hossam Abdelmonem B, Kamal LT, Wardy LW, Ragheb M, Hanna MM, Elsharkawy M, Abdelnaser A. Non-coding RNAs: emerging biomarkers and therapeutic targets in cancer and inflammatory diseases. Front Oncol 2025; 15:1534862. [PMID: 40129920 PMCID: PMC11931079 DOI: 10.3389/fonc.2025.1534862] [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: 11/26/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Non-coding RNAs (ncRNAs) have a significant role in gene regulation, especially in cancer and inflammatory diseases. ncRNAs, such as microRNA, long non-coding RNAs, and circular RNAs, alter the transcriptional, post-transcriptional, and epigenetic gene expression levels. These molecules act as biomarkers and possible therapeutic targets because aberrant ncRNA expression has been directly connected to tumor progression, metastasis, and response to therapy in cancer research. ncRNAs' interactions with multiple cellular pathways, including MAPK, Wnt, and PI3K/AKT/mTOR, impact cellular processes like proliferation, apoptosis, and immune responses. The potential of RNA-based therapeutics, such as anti-microRNA and microRNA mimics, to restore normal gene expression is being actively studied. Additionally, the tissue-specific expression patterns of ncRNAs offer unique opportunities for targeted therapy. Specificity, stability, and immune responses are obstacles to the therapeutic use of ncRNAs; however, novel strategies, such as modified oligonucleotides and targeted delivery systems, are being developed. ncRNA profiling may result in more individualized and successful treatments as precision medicine advances, improving patient outcomes and creating early diagnosis and monitoring opportunities. The current review aims to investigate the roles of ncRNAs as potential biomarkers and therapeutic targets in cancer and inflammatory diseases, focusing on their mechanisms in gene regulation and their implications for non-invasive diagnostics and targeted therapies. A comprehensive literature review was conducted using PubMed and Google Scholar, focusing on research published between 2014 and 2025. Studies were selected based on rigorous inclusion criteria, including peer-reviewed status and relevance to ncRNA roles in cancer and inflammatory diseases. Non-English, non-peer-reviewed, and inconclusive studies were excluded. This approach ensures that the findings presented are based on high-quality and relevant sources.
Collapse
Affiliation(s)
- Basma Hossam Abdelmonem
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
- Basic Sciences Department, Faculty of Physical Therapy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Lereen T. Kamal
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
| | - Lilian Waheed Wardy
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
- Research and Development Department, Eva Pharma for Pharmaceuticals Industries, Cairo, Egypt
| | - Manon Ragheb
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
- School of Medicine, New Giza University (NGU), Giza, Egypt
| | - Mireille M. Hanna
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
| | - Mohamed Elsharkawy
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
| |
Collapse
|
14
|
Gu W, Tian Q, Xie T, Chen X. Genetically reprogrammed exosomes for cancer immunotherapy. Mol Ther 2025; 33:837-839. [PMID: 40010337 PMCID: PMC11897764 DOI: 10.1016/j.ymthe.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Affiliation(s)
- Wenxing Gu
- School of Pharmacy, Hangzhou Normal University, Zhejiang 311121, China; Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore; Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Qingchang Tian
- School of Pharmacy, Hangzhou Normal University, Zhejiang 311121, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Zhejiang 311121, China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore; Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore; Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore 117544, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 138667, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A∗STAR), Singapore 138673, Singapore.
| |
Collapse
|
15
|
Wang F, Wang C, Chen S, Wei C, Ji J, Liu Y, Liang L, Chen Y, Li X, Zhao L, Shi X, Fang Y, Lu W, Li T, Liu Z, Lu W, Li T, Hu X, Li M, Liu F, He X, Wen J, Wang Z, Zhou W, Chen Z, Hong Y, Zhang S, Li X, Zhou R, Mo L, Zhang D, Li T, Zhang Q, Wang L, Wei X, Yang B, Huang S, Zhang H, Pang G, Ouyang L, Wang Z, Cheng J, Xu B, Mo Z. Identification of blood-derived exosomal tumor RNA signatures as noninvasive diagnostic biomarkers for multi-cancer: a multi-phase, multi-center study. Mol Cancer 2025; 24:60. [PMID: 40025576 PMCID: PMC11871737 DOI: 10.1186/s12943-025-02271-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Cancer remains a leading global cause of mortality, making early detection crucial for improving survival outcomes. The study aims to develop a machine learning-enabled blood-derived exosomal RNA profiling platform for multi-cancer detection and localization. METHODS In this multi-phase, multi-center study, we analyzed RNA from exosomes derived from peripheral blood plasma in 818 participants across eight cancer types during the discovery phase. Machine learning techniques were applied to identify potential pan-cancer biomarkers. During the screening and model validation phases, the sample size was progressively expanded to 1,385 participants in two steps, while the candidate biomarkers were refined into a set of 12 exosomal tumor RNA signatures (ETR.sig). In the subsequent model construction phase, diagnostic models were developed using the expanded cohort and ETR.sig. Statistical analyses included the calculation of receiver operating characteristic (ROC) curves and AUC values to assess the models' ability to distinguish cancer cases from controls and determine tumor origins. To further validate and explore the biological relevance of the identified biomarkers, we integrated tissue RNA-seq, single-cell data, and clinical information. RESULTS Machine learning analysis initially identified 33 candidate biomarkers, which were narrowed down to 20 ETR.sig in the screening phase and 12 ETR.sig in the validation phase. In the model construction phase, a diagnostic model based on ETR.sig, built using the Random Forest (RF) algorithm, showed excellent performance with an AUC of 0.915 for distinguishing pan-cancer from controls. The multi-class classification model also demonstrated strong classification power, with macro-average and micro-average AUCs of 0.983 and 0.985, respectively, for differentiating between eight cancer types. Additionally, tumor origin classification using the RF-based diagnostic models achieved high AUC values: BRCA 0.976, COAD 0.98, KIRC 0.947, LIHC 0.967, LUAD 0.853, OV 0.972, PAAD 0.977, and PRAD 0.898. Integration of tissue RNA-seq, single-cell data, and clinical information revealed key associations between ETR.sig-related genes and tumor development. CONCLUSIONS The study demonstrates the robust potential of exosomal RNA as a minimally invasive biomarker resource for cancer detection. The developed ETR.sig platform offers a promising tool for precision oncology and broad-spectrum cancer screening, integrating advanced computational models with nanoscale vesicle biology for accurate and rapid diagnosis.
Collapse
Affiliation(s)
- Fubo Wang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China.
- School of Life Sciences, Guangxi Medical University, Nanning , Guangxi, 530021, China.
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.
- School of Public Health, Guangxi Medical University, Nanning , Guangxi, 530021, China.
| | - Chengbang Wang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China
- Department of Urology, Shanghai Ninth People'S Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Shaohua Chen
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- School of Public Health, Guangxi Medical University, Nanning , Guangxi, 530021, China
| | - Chunmeng Wei
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China
| | - Jin Ji
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Urology, Naval Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Yan Liu
- Department of Breast, Bone and Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, Nanning , Guangxi, 530021, China
- Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi, Department of Education, Affiliated Tumor Hospital of Guangxi Medical University, Nanningaq , Guangxi, 530021, China
| | - Leifeng Liang
- Department of Oncology, The First People'S Hospital of Yulin, the, Sixth Affiliated Hospital of Guangxi Medical Universityaq, Guangxi, 537000, China
| | - Yifeng Chen
- Department of Urology, The First People'S Hospital of Yulin, the, Sixth Affiliated Hospital of Guangxi Medical Universityaq, Guangxi, 537000, China
| | - Xing Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Lin Zhao
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xiaolei Shi
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yu Fang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Weimin Lu
- Department of Urology, Suzhou Hospital of Anhui Medical University, Suzhouaq , AnHui, 234000, China
| | - Tianman Li
- Department of Hepatobiliary Surgery, The First People'S Hospital of Yulin, the, Sixth Affiliated Hospital of Guangxi Medical Universityaq, Guangxi, 537000, China
| | - Zhe Liu
- Department of Urology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Wenhao Lu
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning , Guangxi, 530021, China
| | - Tingting Li
- Department of Breast, Bone and Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, Nanning , Guangxi, 530021, China
- Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi, Department of Education, Affiliated Tumor Hospital of Guangxi Medical University, Nanningaq , Guangxi, 530021, China
| | - Xiangui Hu
- Department of Hepatobiliary and Pancreatic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Mugan Li
- Department of Colorectal and Anal Surgery, The First People'S Hospital of Yulin, the, Sixth Affiliated Hospital of Guangxi Medical Universityaq, Guangxi, 537000, China
| | - Fuchen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Xing He
- Outpatient Department, Qingdao, Special Servicemen Recuperation Center of PLA Navy , Shandong, 266071, China
| | - Jiannan Wen
- The First Outpatient Department, General Hospital of PLA Northern Theater Command, Shenyangaq , Liaoning, 110001, China
| | - Zuheng Wang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China
| | - Wenxuan Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Zehui Chen
- Department of Laboratory Medicine, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China
| | - Yonggang Hong
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Shaohua Zhang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xiao Li
- School of Life Sciences, Guangxi Medical University, Nanning , Guangxi, 530021, China
| | - Rongbin Zhou
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning , Guangxi, 530021, China
| | - Linjian Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China
| | - Duobing Zhang
- Department of Urology, Suzhou Hospital of Anhui Medical University, Suzhouaq , AnHui, 234000, China
- Suzhou Key Laboratory for Clinical Big Data and Intelligent Treatment of Urinary System Diseases, Suzhouaq , AnHui, 234000, China
| | - Tianyu Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China
| | - Qingyun Zhang
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Li Wang
- Research Center for Intelligence Information Technology, Nantong University, Nantong , Jiangsu, 226001, China
| | - Xuedong Wei
- Department of Urology, The First Afliated Hospital of Soochow University, Suzhou, 215006, China
| | - Bo Yang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Shenglin Huang
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 201321, China
| | - Huiyong Zhang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Guijian Pang
- Department of Urology, The First People'S Hospital of Yulin, the, Sixth Affiliated Hospital of Guangxi Medical Universityaq, Guangxi, 537000, China
| | - Liu Ouyang
- Department of Hepatobiliary and Pancreatic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- Department of Hepatobiliary and Pancreatic Surgery, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, 200434, China.
| | - Zhenguang Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| | - Jiwen Cheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People'S Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China.
| |
Collapse
|
16
|
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
|
17
|
Oosterwijk-Wakka JC, Houkes L, van der Zanden LFM, Kiemeney LALM, Junker K, Warren AY, Eisen T, Jaehde U, Radu MT, Ruijtenbeek R, Oosterwijk E. Kinomic profiling to predict sunitinib response of patients with metastasized clear cell Renal Cell Carcinoma. Neoplasia 2025; 60:101108. [PMID: 39724752 PMCID: PMC11732189 DOI: 10.1016/j.neo.2024.101108] [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: 07/23/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Treatment with Sunitinib, a potent multitargeted receptor tyrosine kinase inhibitor (TKI) has increased the progression-free survival (PFS) and overall-survival (OS) of patients with metastasized renal cell carcinoma (mRCC). With modest OS improvement and variable response and toxicity predictive and/or prognostic biomarkers are needed to personalize patient management: Prediction of individual TKI therapy response and resistance will increase successful treatment outcome while reducing unnecessary drug use and expense. The aim of this study was to investigate whether kinase activity analysis can predict sunitinib response and/or toxicity using tissue samples obtained from primary clear cell RCC (ccRCC) from a cohort of clinically annotated patients with mRCC receiving sunitinib as first-line treatment. MATERIALS AND METHODS EuroTARGET partners collected ccRCC and matched normal kidney tissue samples immediately after surgery, snap-frozen and stored at -80°C until use. Phosphotyrosine-activity profiling was performed using PamChip® peptide microarrays (144 peptides derived from known phosphorylation sites in Protein Tyrosine Kinase substrates) of lysed tissue samples (5 µg protein input) of 163 mRCC patients. Evolve software Was used to analyze kinome profiles and Bionavigator was used for unsupervised and supervised clustering. The kinexus kinase predictor (www.phosphonet.ca) was used to analyze the peptide lists within the clusters. RESULTS Kinome data was available from 94 patients who received sunitinib as 1st-line treatment and had complete follow-up of their clinical data (PFS, OS and toxicity) for at least 6 months. Matched normal tissue was available from 14 mRCC patients. Supervised clustering of basal kinome activity could correctly classify mRCC patients with PFS >9 months versus PFS<9 months with an accuracy of 61 %. Unsupervised hierarchical clustering revealed 3 major clusters related to immune signaling, VEGF pathway, and immune signaling/cell adhesion. Basal kinase activity levels of patients with short PFS were substantially higher compared to patients who experienced extended PFS. DISCUSSION/CONCLUSION Based on kinase levels ccRCC tumors can be subdivided into 3 clusters which may reflect the aggressiveness of these tumors. The accuracy of response prediction of 61 % based on basal kinase levels is too low to justify implementation. STK assays may help to predict sunitinib toxicity and guide clinical management. Additionally, it is possible that mRCC patients with an immune kinase signature are better checkpoint inhibitor candidates, but this needs to be studied.
Collapse
Affiliation(s)
| | - Liesbeth Houkes
- PamGene International B.V., 5211 HH 's-Hertogenbosch, the Netherlands
| | | | | | - Kerstin Junker
- Clinic of Urology and Paediatric Urology, Saarland University, 66424 Homburg, Germany
| | - Anne Y Warren
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust and Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Tim Eisen
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust and Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Ulrich Jaehde
- CESAR Central Office, CESAR Central European Society for Anticancer Drug Research-EWIV, 1010, Vienna, Austria
| | - Marius T Radu
- University of Medicine and Pharmacy Carol Davila 050474, Bucharest, Romania
| | - Rob Ruijtenbeek
- PamGene International B.V., 5211 HH 's-Hertogenbosch, the Netherlands
| | - Egbert Oosterwijk
- Radboud University Medical Center, 6525 GA, Nijmegen, the Netherlands
| |
Collapse
|
18
|
Xiao H, Fei M, Xu Q, Gao Y, Feng R, Liang C, Wang B, Li H. MATN1-AS1 Promotes Tumour Metastasis and Sunitinib Resistance via E2F2 in Clear Cell Renal Cell Carcinoma. J Cell Mol Med 2025; 29:e70428. [PMID: 39999286 PMCID: PMC11855375 DOI: 10.1111/jcmm.70428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/14/2024] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
It has become increasingly recognised that MATN1-AS1 is involved in multiple tumour development. The role of MATN1-AS1 in clear cell renal cell carcinoma (ccRCC), however, is still largely unrecognised. This study investigated the molecular functions of MATN1-AS1 in promoting ccRCC metastasis and sunitinib resistance. MATN1-AS1 was found to be mainly located in the cytoplasm and was upregulated in ccRCC, and a positive association was seen between greater levels of MATN1-AS1 expression and worse clinical outcomes. Downregulating MATN1-AS1 significantly hindered cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT). MATN1-AS1 promoted tumour growth and metastasis in vivo. Mechanismly, MATN1-AS1 targeted microRNA miR-214-5p, thereby upregulating E2F2 and promoting E2F2-mediated EMT. We discovered that MATN1-AS1 also promoted sunitinib resistance via E2F2 in vitro. Collectively, our research uncovered the protumor characteristics of MATN1-AS1 and suggested it as a therapeutic target for reverse sunitinib resistance in ccRCC.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Sunitinib/pharmacology
- Sunitinib/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/metabolism
- Epithelial-Mesenchymal Transition/genetics
- Epithelial-Mesenchymal Transition/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Line, Tumor
- Animals
- Cell Movement/genetics
- Cell Movement/drug effects
- Cell Proliferation/genetics
- Cell Proliferation/drug effects
- E2F2 Transcription Factor/genetics
- E2F2 Transcription Factor/metabolism
- Mice
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Neoplasm Metastasis
- Male
- Female
- MicroRNAs/genetics
- Mice, Nude
Collapse
Affiliation(s)
- Haibing Xiao
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Mintian Fei
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Qili Xu
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Yu Gao
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Rui Feng
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
- Department of UrologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chaozhao Liang
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Baojun Wang
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
| | - Haolin Li
- Anhui Provincal Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityAnhuiChina
- Department of UrologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Khan Y, Hussain MS, Ramalingam PS, Fatima R, Maqbool M, Ashique S, Khan NU, Bisht AS, Gupta G. Exploring extracellular RNA as drivers of chemotherapy resistance in cancer. Mol Biol Rep 2025; 52:142. [PMID: 39836259 DOI: 10.1007/s11033-025-10263-2] [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] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Chemotherapy resistance (CR) represents one of the most important barriers to effective oncological therapy and often leads to ineffective intervention and unfavorable clinical prognosis. Emerging studies have emphasized the vital significance of extracellular RNA (exRNA) in influencing CR. This thorough assessment intends to explore the multifaceted contributions of exRNA, such as exosomal RNA, microRNAs, long non-coding RNAs, and circular RNAs, to CR in cancer. We discuss the mechanisms by which exRNA facilitates drug resistance, such as modulating gene expression, influencing the tumor microenvironment, and facilitating intercellular communication. Furthermore, we examine the potential of exRNA as prognostic factor for determining oncology treatment efficacy and their emerging role as therapeutic targets. Diagnostic and prognostic applications of exRNA biomarkers are considered, alongside current methodologies for their detection and quantification. Additionally, we review recent advances in exRNA-targeted therapies, highlighting ongoing clinical trials and therapeutic strategies aimed at overcoming chemoresistance. Despite the promise of exRNA research, several challenges remain, including technical limitations and the biological complexity of exRNA networks. This review underscores the importance of continued investigation into exRNA biology and its therapeutic potential, which in the future may provide new avenues for cancer treatment and tailored medical strategies. By elucidating the role of exRNA in CR, this article aims to provide a comprehensive resource for researchers and clinicians seeking to improve the effectiveness of carcinoma management approaches.
Collapse
Affiliation(s)
- Yumna Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, PO Box 25130, Pakistan
| | - Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248007, India.
| | - Prasanna Srinivasan Ramalingam
- Protein Engineering Lab, School of Biosciences and Technology, Vellore Institute of Technology, Katpadi, Vellore, Tamil Nadu, 632014, India
| | - Rabab Fatima
- Department of Chemistry, Energy Acres, University of Petroleum & Energy Studies, Dehradun, Uttarakhand, 248007, India
| | - Mudasir Maqbool
- Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Jammu, Srinagar, Kashmir, 190006, India
| | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, PO Box 25130, Pakistan
| | - Ajay Singh Bisht
- School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Patel Nagar, Dehradun, Uttarakhand, 248001, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Punjab, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, UAE
| |
Collapse
|
22
|
Huang S, Qin X, Fu S, Hu J, Jiang Z, Hu M, Zhang B, Liu J, Chen Y, Wang M, Liu X, Chen Z, Wang L. STAMBPL1/TRIM21 Balances AXL Stability Impacting Mesenchymal Phenotype and Immune Response in KIRC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405083. [PMID: 39527690 PMCID: PMC11714167 DOI: 10.1002/advs.202405083] [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: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Kidney renal clear cell carcinoma (KIRC) is recognized as an immunogenic tumor, and immunotherapy is incorporated into its treatment landscape for decades. The acquisition of a tumor mesenchymal phenotype through epithelial-to-mesenchymal transition (EMT) is associated with immune evasion and can contribute to immunotherapy resistance. Here, the involvement of STAM Binding Protein Like 1 (STAMBPL1) is reported in the development of mesenchymal and immune evasion phenotypes in KIRC cells. Mechanistically, STAMBPL1 elevated protein abundance and surface accumulation of TAM Receptor AXL through diminishing the TRIM21-mediated K63-linked ubiquitination and subsequent lysosomal degradation of AXL, thereby enhancing the expression of mesenchymal genes while suppressing chemokines CXCL9/10 and HLA/B/C. In addition, STAMBPL1 enhanced PD-L1 transcription via facilitating nuclear translocation of p65, and knockdown (KD) of STAMBPL1 augmented antitumor effects of PD-1 blockade. Furthermore, STAMBPL1 silencing and the tyrosine kinase inhibitor (TKI) sunitinib also exhibited a synergistic effect on the suppression of KIRC. Collectively, targeting the STAMBPL1/TRIM21/AXL axis can decrease mesenchymal phenotype and potentiate anti-tumor efficacy of cancer therapy.
Collapse
Affiliation(s)
- Shiyu Huang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Xuke Qin
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Shujie Fu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Juncheng Hu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Zhengyu Jiang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Min Hu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Banghua Zhang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Hubei Key Laboratory of Digestive System DiseaseWuhan430060China
| | - Jiachen Liu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Yujie Chen
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Minghui Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Xiuheng Liu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Zhiyuan Chen
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Lei Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| |
Collapse
|
23
|
Wu J, Zhang C, Li H, Zhang S, Chen J, Qin L. Competing endogenous RNAs network dysregulation in oral cancer: a multifaceted perspective on crosstalk and competition. Cancer Cell Int 2024; 24:431. [PMID: 39725978 DOI: 10.1186/s12935-024-03580-2] [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: 04/26/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
Oral cancer progresses from asymptomatic to advanced stages, often involving cervical lymph node metastasis, resistance to chemotherapy, and an unfavorable prognosis. Clarifying its potential mechanisms is vital for developing effective theraputic strategies. Recent research suggests a substantial involvement of non-coding RNA (ncRNA) in the initiation and advancement of oral cancer. However, the underlying roles and functions of various ncRNA types in the growth of this malignant tumor remain unclear. Competing endogenous RNAs (ceRNAs) refer to transcripts that can mutually regulate each other at the post-transcriptional level by vying for shared miRNAs. Networks of ceRNAs establish connections between the functions of protein-coding mRNAs and non-coding RNAs, including microRNA, long non-coding RNA, pseudogenic RNA, and circular RNA, piwi-RNA, snoRNA. A growing body of research has indicated that imbalances in ceRNAs networks play a crucial role in various facets of oral cancer, including development, metastasis, migration, invasion, and inflammatory responses. Hence, delving into the regulatory pathways of ceRNAs in oral cancer holds the potential to advance our understanding of the pathological mechanisms, facilitate early diagnosis, and foster targeted drug development for this malignancy. The present review summarized the fundamental role of ceRNA network, discussed the limitations of current ceRNA applications, which have been improved through chemical modification and carrier delivery as new biomarkers for diagnosis and prognosis is expected to offer a groundbreaking therapeutic approach for individuals with oral cancer.
Collapse
Affiliation(s)
- Jiajun Wu
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Chanjuan Zhang
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Hongfang Li
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Shuo Zhang
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Jingxin Chen
- Department of Oral and Maxillofacial Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, China.
- School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, Changsha, Hunan, 410208, China.
| | - Li Qin
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
24
|
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
|
25
|
Alkhathami AG, Pallathadka H, Shah S, Ganesan S, Sharma A, Devi S, Mustafa YF, Alasheqi MQ, Kadhim AJ, Zwamel AH. LncRNAs in modulating cancer cell resistance to paclitaxel (PTX) therapy. Med Oncol 2024; 42:28. [PMID: 39671022 DOI: 10.1007/s12032-024-02577-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
Paclitaxel (PTX) is widely used for treating several cancers, including breast, ovarian, lung, esophageal, gastric, pancreatic, and neck cancers. Despite its clinical utility, cancer recurrence frequently occurs in patients due to the development of resistance to PTX. Resistance mechanisms in cancer cells treated with PTX include alterations in β-tubulin, the target molecule involved in mitosis, activation of molecular pathways enabling drug efflux, and dysregulation of apoptosis-related proteins. Long non-coding RNAs (lncRNAs), which are RNA molecules longer than 200 nucleotides without protein-coding potential, serve diverse regulatory roles in cellular processes. Increasing evidence highlights the involvement of lncRNAs in cancer progression and their contribution to PTX resistance across various cancers. Consequently, lncRNAs have emerged as potential therapeutic targets for addressing drug resistance in cancer treatment. This review focuses on the current understanding of lncRNAs and their role in drug resistance mechanisms, aiming to encourage further investigation in this area. Key lncRNAs and their associated pathways linked to PTX resistance will be summarized.
Collapse
Affiliation(s)
- Ali G Alkhathami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Sejal Shah
- Department of Bioinformatics, Faculty of Engineering and Technology, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhishek Sharma
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Seema Devi
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab, 140307, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
26
|
Wang S, Qi X, Liu D, Xie D, Jiang B, Wang J, Wang X, Wu G. The implications for urological malignancies of non-coding RNAs in the the tumor microenvironment. Comput Struct Biotechnol J 2024; 23:491-505. [PMID: 38249783 PMCID: PMC10796827 DOI: 10.1016/j.csbj.2023.12.016] [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: 10/03/2023] [Revised: 12/08/2023] [Accepted: 12/16/2023] [Indexed: 01/23/2024] Open
Abstract
Urological malignancies are a major global health issue because of their complexity and the wide range of ways they affect patients. There's a growing need for in-depth research into these cancers, especially at the molecular level. Recent studies have highlighted the importance of non-coding RNAs (ncRNAs) – these don't code for proteins but are crucial in controlling genes – and the tumor microenvironment (TME), which is no longer seen as just a background factor but as an active player in cancer progression. Understanding how ncRNAs and the TME interact is key for finding new ways to diagnose and predict outcomes in urological cancers, and for developing new treatments. This article reviews the basic features of ncRNAs and goes into detail about their various roles in the TME, focusing specifically on how different ncRNAs function and act in urological malignancies.
Collapse
Affiliation(s)
- Shijin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Xiaochen Qi
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Deqian Xie
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Bowen Jiang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Jin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Xiaoxi Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| |
Collapse
|
27
|
Jiang A, Liu Y, He Z, Liu W, Yang Q, Fang Y, Zhu B, Wu X, Ye H, Ye B, Gao S, Qu L, Xu W, Luo P, Wang L. TDERS, an exosome RNA-derived signature predicts prognosis and immunotherapeutic response in clear cell renal cell cancer: a multicohort study. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:382-394. [PMID: 39735439 PMCID: PMC11674438 DOI: 10.1016/j.jncc.2024.07.002] [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: 09/05/2023] [Revised: 06/08/2024] [Accepted: 07/23/2024] [Indexed: 12/31/2024] Open
Abstract
Background Tumor-derived exosomes are involved in tumor progression and immune invasion and might function as promising noninvasive approaches for clinical management. However, there are few reports on exosom-based markers for predicting the progression and adjuvant therapy response rate among patients with clear cell renal cell carcinoma (ccRCC). Methods The signatures differentially expressed in exosomes from tumor and normal tissues from ccRCC patients were correspondingly deregulated in ccRCC tissues. We adopted a two-step strategy, including Lasso and bootstrapping, to construct a novel risk stratification system termed the TDERS (Tumor-Derived Exosome-Related Risk Score). During the testing and validation phases, we leveraged multiple external datasets containing over 2000 RCC cases from eight cohorts and one inhouse cohort to evaluate the accuracy of the TDERS. In addition, enrichment analysis, immune infiltration signatures, mutation landscape and therapy sensitivity between the high and low TDERS groups were compared. Finally, the impact of TDERS on the tumor microenvironment (TME) was also analysed in our single-cell datasets. Results TDERS consisted of 12 mRNAs deregulated in both exosomes and tissues from patients with ccRCC. TDERS achieved satisfactory performance in both prognosis and immune checkpoint inhibitor (ICI) response across all ccRCC cohorts and other pathological types, since the average area under the curve (AUC) to predict 5-year overall survival (OS) was larger than 0.8 across the four cohorts. Patients in the TDERS high group were resistant to ICIs, while mercaptopurine might function as a promising agent for those patients. Patients with a high TDERS were characterized by coagulation and hypoxia, which induced hampered tumor antigen presentation and relative resistance to ICIs. In addition, single cells from 12 advanced samples validated this phenomenon since the interaction between dendritic cells and macrophages was limited. Finally, PLOD2, which is highly expressed in fibro- and epi‑tissue, could be a potential therapeutic target for ccRCC patients since inhibiting PLOD2 altered the malignant phenotype of ccRCC in vitro. Conclusion As a novel, non-invasive, and repeatable monitoring tool, the TDERS could work as a robust risk stratification system for patients with ccRCC and precisely inform treatment decisions about ICI therapy.
Collapse
Affiliation(s)
- Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ziwei He
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qiwei Yang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Urology, the Third Affiliated Hospital of Naval Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Yu Fang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Baohua Zhu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xiaofeng Wu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Huamao Ye
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Bicheng Ye
- School of Clinical Medicine, Medical College of Yangzhou Polytechnic College, Yangzhou, China
| | - Shunxiang Gao
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
28
|
Lin A, Li J, He W. CircSLC4A7 in resistant-cells-derived exosomes promotes docetaxel resistance via the miR-1205/MAPT axis in prostate cancer. IUBMB Life 2024; 76:1342-1355. [PMID: 39266461 DOI: 10.1002/iub.2915] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/06/2024] [Indexed: 09/14/2024]
Abstract
Prostate cancer (PCa) is a high-mortality cancer. Docetaxel (DCT) combined with second-generation anti-androgens is considered the golden standard therapy for PCa, whose application is limited for DCT resistance (DR). Therefore, exploring the mechanism of DR is of great importance. In this study, PCa cell lines of PC3 and DU145 were employed, and DR cells were constructed by treatment with graded DCT. CircSLC4A7, miR-1205, and microtubule-associated protein tau (MAPT) transfections were established. Cell counting kit-8 assay was performed to evaluate the cell activity and IC50 of DCT. After being treated with DCT, DR was assessed by colony formation assay, flow cytometry analysis, and terminal transferase-mediated UTP nick end-labeling assay. Real-time quantitative PCR and western blotting analysis evaluated the expression levels of genes. The dual-luciferase reporter gene assay verified the miR-1205 binding sites with circSLC4A7 and MAPT. An animal experiment was performed to assess the tumor growth influenced by circSLC4A7. After conducting DR cells and isolated exosomes, we found that not only co-culture with DR cells but also treatment with DR cells' exosomes would promote the DR of normal cells. Moreover, circSLC4A7 was highly expressed in DR cells and their exosomes. CircSLC4A7 overexpression enhanced DR, represented as raised IC50 of DCT, increased colony formation, and decreased cell apoptosis after DCT treatment, while circSLC4A7 knockdown had the opposite effect. MiR-1205 was confirmed as a circSLC4A7-sponged miRNA and miR-1205 inhibitor reversed the effect of sh-circSLC4A7. MAPT was further identified as a target of miR-1205 and had a similar effect with circSLC4A7. The effect of circSLC4A7 on DR was also confirmed by xenograft experiments. Collectively, circSLC4A7 in resistant-cells-derived exosomes promotes DCT resistance of PCa via miR-1205/MAPT axis, which may provide a new treatment strategy for DR of PCa.
Collapse
Affiliation(s)
- Anhua Lin
- Department of Endocrinology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Junhe Li
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenjing He
- Department of Endocrinology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
29
|
Tian X, Liu J, Yi C, You X, Yuan C. Hsa_circ_0072732 enhances sunitinib resistance of renal cell carcinoma by inhibiting ferroptosis. Discov Oncol 2024; 15:700. [PMID: 39580569 PMCID: PMC11585529 DOI: 10.1007/s12672-024-01580-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is one of the most diagnosed urological malignancies with high mortality and increasing incidence. What's more, the sunitinib resistance undoubtedly increased the difficulties in RCC therapy. Circular RNAs (circRNAs) are a newly found type of non-coding RNAs with a special circular structure, and are found to participate in the occurrence development, chemoresistance, and prognosis of cancers. Ferroptosis regulates disease progression mainly via polyunsaturated fatty acid metabolism and glutamine catabolic pathways. The mechanism of circRNAs contributed to sunitinib resistance through ferroptosis has not been elucidated clearly. MATERIALS AND METHODS In our research, we identified a novel circRNA Hsa_circ_0072732 from circRNA datasets (GSE108735 and GSE100186). RNase R and Actinomycin D assays were used to detect the loop structure and stability of circRNAs. qRT-PCR and western blot were used for the detection of RNA and protein levels. CCK8 assays were used to detect proliferation and cell viability. Lipid peroxidation (MDA), and reactive oxygen species (ROS) were detected by indicted kits. Dual-luciferase reporter and RNA pull-down assays were used to detect the RNA interactions. RESULTS Our results showed that Hsa_circ_0072732 was highly expressed in RCC cells. Further investigations showed that the silence of Hsa_circ_0072732 could increase RCC sensitivity to sunitinib. Hsa_circ_0072732 contributed to sunitinib chemoresistance by impairing ferroptosis. Hsa_circ_0072732 exerts its function mainly by acting as sponges for miR-548b-3p and regulating the expression SLC7A11. Our research suggests that ferroptosis is involved in sunitinib resistance, and targeting ferroptosis is a promising way for RCC treatment. CONCLUSION Our research suggests Hsa_circ_0072732 enhanced renal cell carcinoma sunitinib resistance by inhibiting ferroptosis through miR-548b-3p/SLC7A11.
Collapse
Affiliation(s)
- Xiaorui Tian
- Department of Urology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443002, China
- Department of Urology, Yichang Central People's Hospital, Yichang, 443002, China
| | - Jun Liu
- Nursing Department, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Cheng Yi
- Department of Urology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Xiangyun You
- Department of Urology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Chunli Yuan
- Department of Urology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443002, China.
| |
Collapse
|
30
|
Sui X, Zhang Q, Hao M, Chen Y. Serum LINC01133 combined with CEA and CA19-9 contributes to the diagnosis and survival prognosis of gastric cancer. Medicine (Baltimore) 2024; 103:e40564. [PMID: 39560546 PMCID: PMC11575958 DOI: 10.1097/md.0000000000040564] [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: 12/05/2023] [Accepted: 10/29/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) are currently 2 major diagnostic biomarkers for gastric cancer (GC). The aims of study were to detect the expression of long intergenic nonprotein coding RNA 1133 (LINC01133), and to evaluate its diagnostic and prognostic value in GC. Furthermore, the clinical performance of the joint detection of LINC01133, CEA and CA19-9 was also evaluate in GC. METHODS The data were collected from 156 GC, 96 chronic superficial gastritis, 77 chronic atrophic gastritis patients and 89 healthy controls. LINC01133 expression was determined by quantitative real-time PCR. Receiver operating characteristics analysis was used to evaluate the diagnostic value of LINC01133, CEA, CA19-9 individually and jointly. Kaplan-Meier method and log-rank test were used to conduct survival comparison analysis. Cox regression was used to screen the independent prognostic factors for GC. RESULTS Serum LINC01133 expression was decreased in GC patients compared with chronic superficial gastritis, chronic atrophic gastritis and healthy controls, and had considerable diagnostic potential, and notably, the joint detection of LINC01133, CEA, and CA19-9 showed the highest diagnostic accuracy in distinguishing GC patients from healthy or gastritis patients. LINC01133 expression was associated with GC patients' CEA and CA19-9 levels, tumor size, differentiation, lymph node metastasis and tumor node metastasis stage. Low LINC01133 was associated with poor GC survival, and was an independent prognostic factor for GC. CONCLUSION Decreased serum LINC01133 had considerable diagnostic potential, and the joint detection of LINC01133, CEA, and CA19-9 might be a more efficient diagnostic strategy for GC patients. Reduced LINC01133 served as a prognostic biomarker to predict poor GC survival.
Collapse
Affiliation(s)
- Xiaomei Sui
- Department of Radiotherapy, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Qifu Zhang
- Department of Radiotherapy, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Meili Hao
- Department of Radiotherapy, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yanfang Chen
- Department of Radiotherapy, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
31
|
Jia M, Xie M, Luo X, Wang H, Duan C, Lai W, Dai R, Wang R. Cancer-Associated Fibroblast-Derived FGF7 Promotes Clear Cell Renal Cell Carcinoma Progression and Macrophage Infiltration. Cells 2024; 13:1824. [PMID: 39594574 PMCID: PMC11593278 DOI: 10.3390/cells13221824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/18/2024] [Accepted: 10/27/2024] [Indexed: 11/28/2024] Open
Abstract
As the predominant stromal cells in the ccRCC surrounding environment, cancer-associated fibroblasts (CAFs) have been established as supportive of tumor growth. However, the detailed molecular mechanisms underlying the supporting role of CAFs in ccRCC have not been well characterized. Evidence from the clustering consensus analysis, single-cell analysis, and the experimental results illustrate that CAF-derived FGF7 plays a crucial role as a signaling mediator between CAFs and ccRCC tumor cells. Mechanistically, CAF-derived FGF7 triggers AKT activation to promote cell growth and cell invasion of ccRCC tumor cells. As a response, ccRCC tumor cells stimulate STAT3-mediated transcriptional regulation, directly increasing FGF7 expression at the chromatin level in CAFs. Moreover, there exists a positive clinical correlation between the abundance of CAFs, FGF7 expression, and the infiltration of M2 type macrophages. The RENCA in vivo mouse model also confirmed that FGF7 depletion could impede RCC development by reducing the recruitment of M2 type macrophages. Overall, this study delineates a key signaling axis governing the crosstalk between CAFs and ccRCC tumor cells, highlighting FGF7 as a promising therapeutic target of ccRCC.
Collapse
Affiliation(s)
- Man Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Mingyu Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Xixi Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Huiping Wang
- Department of Genetics, Xuzhou Medical University, Xuzhou 221004, China
| | - Chunyan Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Wanni Lai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Rongyang Dai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Ronghao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
32
|
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
|
33
|
Taghavinia F, Akhlaghipour I, Golshan A, Aarabi A, Abbaszadegan MR, Moghbeli M. LINC00365 as a potential biomarker for total nephrectomy in advanced-stage renal cell carcinoma patients. Pathol Res Pract 2024; 263:155630. [PMID: 39353324 DOI: 10.1016/j.prp.2024.155630] [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: 08/23/2024] [Revised: 09/11/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Renal cell carcinoma (RCC) is one of the most frequent urological cancers globally that has a good prognosis in the early tumor stages. However, there is a poor prognosis in metastatic RCC patients. Therefore, it is needed to evaluate the molecular biology of RCC progression to introduce the efficient diagnostic and therapeutic markers in these patients. Long non-coding RNAs (lncRNAs) have key roles in regulation of molecular mechanisms during RCC progression. In the present study, we assessed the levels of LINC00365 expressions in RCC patients to suggest that as a tumor marker in these patients. METHODS Fifty fresh RCC tumor tissues and their normal margins were collected to assess the levels of LINC00365 expressions and probable correlations with clinicopathological features of RCC patients. RESULTS There was significant LINC00365 up regulation in females compared with males (p=0.050). Among the RCC patients with total nephrectomy, there was a significant LINC00365 up regulation in advanced stage compared with primary stage tumors (p=0.035). RCC patients older than 60 years old who were undergone the total nephrectomy had also significant LINC00365 up regulation compared with RCC patients younger than 60 years old (p=0.039). CONCLUSIONS given the significant increase in LINC00365 expression in advanced stage RCC tumors and patients over 60 years old who had total nephrectomy; it could serve as a useful diagnostic marker in screening programs for old high-risk individuals. It was also noticed that female RCC patients had elevated levels of LINC00365 expressions in their tumor samples, suggesting its potential use as a gender-specific diagnostic marker for high-risk females. Nevertheless, evaluating the levels of LINC00365 in serum samples of RCC patients is necessary to suggest that as a reliable diagnostic marker in clinical settings.
Collapse
Affiliation(s)
- Fatemeh Taghavinia
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Golshan
- Department of Urology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Kidney Transplantation Complications Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azadeh Aarabi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
34
|
Meng X, Tan Z, Qiu B, Zhang J, Wang R, Ni W, Fan J. METTL3-induced lncARSR aggravates neuroblastoma tumorigenic properties through stabilizing PHOX2B. Pathol Res Pract 2024; 263:155670. [PMID: 39461245 DOI: 10.1016/j.prp.2024.155670] [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: 05/20/2024] [Revised: 08/25/2024] [Accepted: 10/19/2024] [Indexed: 10/29/2024]
Abstract
Neuroblastoma (NB), the most common extracranial solid tumor in pediatric patients, manifests with considerable variability across multiple primary sites. Despite this, the extent of genetic heterogeneity within these tumor foci and the identification of consistent oncogenic drivers remains largely unexplored. Of particular interest, genetic mutations in PHOX2B have been linked to familial cases of NB, yet the underlying molecular mechanisms are not fully delineated. In our research, we focus on unraveling the role of a novel functional long non-coding RNA (lncRNA) associated with PHOX2B in the context of NB. Using NB cell models with overexpressed PHOX2B, combined with lncRNA microarray analysis, we discovered that lncARSR is significantly upregulated in response to PHOX2B overexpression. Subsequent biological assays demonstrated that lncARSR promotes both the proliferation and metastasis of NB cells. Further molecular investigations revealed that lncARSR plays a crucial role in stabilizing PHOX2B expression within NB cells. Moreover, we identified that the expression of lncARSR is regulated by methylation through methyltransferase-like 3 (METTL3), which itself is positively correlated with PHOX2B expression. Rescue experiments underscored the functional importance of METTL3, lncARSR, and PHOX2B in NB cells. In summary, our findings provide new insights into the molecular functions of PHOX2B in the progression of neuroblastoma and propose a novel therapeutic target for this aggressive malignancy.
Collapse
Affiliation(s)
- Xiangyi Meng
- Department of Pediatrics, Shenzhen University General Hospital, China
| | - Zhu Tan
- Department of Pediatrics, Shenzhen University General Hospital, China
| | - Bihua Qiu
- Department of Pediatrics, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Jie Zhang
- Department of Pediatrics, Shenzhen University General Hospital, China
| | - Ruobing Wang
- Department of Pediatrics, Shenzhen University General Hospital, China
| | - Wensi Ni
- Department of Pediatrics, Shenzhen University General Hospital, China.
| | - Jialing Fan
- Department of Pediatrics, Shenzhen University General Hospital, China.
| |
Collapse
|
35
|
Zhu T, Fu H, Wang Z, Guo S, Zhang S. Identification of exosomal ceRNA networks as prognostic markers in clear cell renal cell carcinoma. Medicine (Baltimore) 2024; 103:e40167. [PMID: 39470474 PMCID: PMC11521039 DOI: 10.1097/md.0000000000040167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024] Open
Abstract
Aggressive clear cell renal cell carcinoma (ccRCC) has a bad prognosis. We seek new ccRCC biomarkers for diagnosis and treatment. We used exoRBase and The Cancer Genome Atlas Database to compare DEmRNAs, DEmiRNAs, DElncRNAs, and DEcircRNAs in ccRCC and normal renal tissues. CircRNAs and circRNAs targeting microRNAs (miRNAs) were anticipated and taken intersections, and several databases assessed the targeted link between common miRNAs and messenger RNAs (mRNAs). The Cancer Genome Atlas database was used to create a predictive mRNA signature that was validated in E-MTAB-1980. Finally, we examined competing endogenous RNA network miRNAs and long noncoding RNAs for ccRCC predictive biomarkers using overall survival analysis. We built the first competing endogenous RNA regulation network of circRNA-lncRNA-miRNA-mRNA and found that it substantially correlates with ccRCC prognosis. We unveiled ccRCC's posttranscriptional regulation mechanism in greater detail. Our findings identified novel biomarkers for ccRCC diagnosis, therapy, and prognosis.
Collapse
Affiliation(s)
- Tao Zhu
- Department of Urology, Weifang People’s Hospital, Weifang, Shandong Province, China
| | - Haizhu Fu
- Department of Urology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong Province, China
| | - Zhiqiang Wang
- Department of Urology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong Province, China
| | - Shanchun Guo
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA
| | - Shidong Zhang
- Department of Urology, Weifang People’s Hospital, Weifang, Shandong Province, China
| |
Collapse
|
36
|
Wang S, Bai Y, Ma J, Qiao L, Zhang M. Long non-coding RNAs: regulators of autophagy and potential biomarkers in therapy resistance and urological cancers. Front Pharmacol 2024; 15:1442227. [PMID: 39512820 PMCID: PMC11540796 DOI: 10.3389/fphar.2024.1442227] [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: 06/01/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
The non-coding RNAs (ncRNAs) comprise a large part of human genome that mainly do not code for proteins. Although ncRNAs were first believed to be non-functional, the more investigations highlighted tthe possibility of ncRNAs in controlling vital biological processes. The length of long non-coding RNAs (lncRNAs) exceeds 200 nucleotidesand can be present in nucleus and cytoplasm. LncRNAs do not translate to proteins and they have been implicated in the regulation of tumorigenesis. On the other hand, One way cells die is by a process called autophagy, which breaks down proteins and other components in the cytoplasm., while the aberrant activation of autophagy allegedly involved in the pathogenesis of diseases. The autophagy exerts anti-cancer activity in pre-cancerous lesions, while it has oncogenic function in advanced stages of cancers. The current overview focuses on the connection between lncRNAs and autophagy in urological cancers is discussed. Notably, one possible role for lncRNAs is as diagnostic and prognostic variablesin urological cancers. The proliferation, metastasis, apoptosis and therapy response in prostate, bladder and renal cancers are regulated by lncRNAs. The changes in autophagy levels can also influence the apoptosis, proliferation and therapy response in urological tumors. Since lncRNAs have modulatory functions, they can affect autophagy mechanism to determine progression of urological cancers.
Collapse
Affiliation(s)
- Shizong Wang
- Department of Urology, Weifang People’s Hospital, Weifang, Shandong, China
- Shangdong Provincial Key Laboratory for Prevention and Treatment of Urological Diseases in Medicine and Health, Weifang, Shandong, China
| | - Yang Bai
- Department of Urology, Weifang People’s Hospital, Weifang, Shandong, China
- Shangdong Provincial Key Laboratory for Prevention and Treatment of Urological Diseases in Medicine and Health, Weifang, Shandong, China
| | - Jie Ma
- Department of Urology, Weifang People’s Hospital, Weifang, Shandong, China
- Shangdong Provincial Key Laboratory for Prevention and Treatment of Urological Diseases in Medicine and Health, Weifang, Shandong, China
| | - Liang Qiao
- Department of Urology, Weifang People’s Hospital, Weifang, Shandong, China
- Shangdong Provincial Key Laboratory for Prevention and Treatment of Urological Diseases in Medicine and Health, Weifang, Shandong, China
| | - Mingqing Zhang
- Department of Urology, Weifang People’s Hospital, Weifang, Shandong, China
- Shangdong Provincial Key Laboratory for Prevention and Treatment of Urological Diseases in Medicine and Health, Weifang, Shandong, China
| |
Collapse
|
37
|
Wang X, Liu T, Li Y, Ding A, Zhang C, Gu Y, Zhao X, Cheng S, Cheng T, Wu S, Duan L, Zhang J, Yin R, Shang M, Gao S. A splicing isoform of PD-1 promotes tumor progression as a potential immune checkpoint. Nat Commun 2024; 15:9114. [PMID: 39438489 PMCID: PMC11496882 DOI: 10.1038/s41467-024-53561-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
The immune checkpoint receptor, programmed cell death 1 (PD-1, encoded by PDCD1), mediates the immune escape of cancer, but whether PD-1 splicing isoforms contribute to this process is still unclear. Here, we identify an alternative splicing isoform of human PD-1, which carries a 28-base pairs extension retained from 5' region of intron 2 (PD-1^28), is expressed in peripheral T cells and tumor infiltrating lymphocytes. PD-1^28 expression is induced on T cells upon activation and is regulated by an RNA binding protein, TAF15. Functionally, PD-1^28 inhibits T cell proliferation, cytokine production, and tumor cell killing in vitro. In vivo, T cell-specific exogenous expression of PD-1^28 promotes tumor growth in both a syngeneic mouse tumor model and humanized NOG mice inoculated with human lung cancer cells. Our study thus demonstrates that PD-1^28 functions as an immune checkpoint, and may contribute to resistance to immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Xuetong Wang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, China
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Tongfeng Liu
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Medical School of Guizhou University, Guiyang, China
| | - Yifei Li
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ao Ding
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chang Zhang
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Department of medical oncology, The Key Laboratory of Advanced Interdisciplinary Studies Center, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangdong, China
| | - Yinmin Gu
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Xujie Zhao
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Shuwen Cheng
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Tianyou Cheng
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Songzhe Wu
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Liqiang Duan
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Jihang Zhang
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Medical School of Guizhou University, Guiyang, China
| | - Rong Yin
- Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Man Shang
- Nanjing Women and Children's Healthcare Institute, Women' s Hospital of Nanjing Medical University (Nanjing Women and Children' s Healthcare Hospital), Nanjing, China
| | - Shan Gao
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, China.
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China.
| |
Collapse
|
38
|
Teng S, Ge J, Yang Y, Cui Z, Min L, Li W, Yang G, Liu K, Wu J. M1 macrophages deliver CASC19 via exosomes to inhibit the proliferation and migration of colon cancer cells. Med Oncol 2024; 41:286. [PMID: 39402192 DOI: 10.1007/s12032-024-02444-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/18/2023] [Indexed: 11/14/2024]
Abstract
Colorectal cancer (CRC) continues to be one of the leading causes of cancer-related death worldwide. Exosomes have been established to play an important role in intercellular communication and that long non-coding RNA (lncRNA) CASC19 is enriched within M1 macrophage-derived exosomes (M1-exo). However, the biological functions and underlying molecular mechanisms of exosomal CASC19 from macrophages on CRC remain unknown. Cell proliferation and migration were evaluated by MTS and transwell assays. The exosomes were characterized by western blot, nanoparticle tracking analysis (NTA) and electron microscope imaging. The expression levels of CASC19 and its putative target miR-410-3p were quantified by reverse-transcription polymerase chain reaction (RT-qPCR). The interaction between CASC19 and miR-410-3p was detected by the pull-down assay. We found that the non-contact inhibition of M1 macrophages on the proliferation of colon cancer cells is largely dependent on the CASC19 released from M1 exosomes. M1 exosomes successfully delivered CASC19 to colon cancer cells, exerting an inhibitory effect on cell proliferation and migration. The exosomes secreted by M1 cells with CASC19 knockdown showed less inhibition effect on cell proliferation and migration. Mechanically, CASC19 exerted an inhibitory effect on colon cancer cells by sponging miR-410-3p via tube morphogenesis and TGF-β signaling pathway. We first proved that CASC19 in M1 macrophages is delivered into colon cancer cells via exosomes, exerting an inhibitory effect on their proliferation and migration by sponging miR-410-3p. The study may provide mechanistic insights into the roles of lncRNAs in CRC progression and a potential therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Shuo Teng
- Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Jiang Ge
- The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
- Qilu Hospital of Shandong University, Qingdao, 266600, China
| | - Yi Yang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Zilu Cui
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Wenkun Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Guodong Yang
- North Sichuan Medical College, Nanchong, 637000, China
| | - Kuiliang Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China.
| | - Jing Wu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China.
- Department of Gastroenterology, Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China.
| |
Collapse
|
39
|
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
|
40
|
Di Bella MA, Taverna S. Extracellular Vesicles: Diagnostic and Therapeutic Applications in Cancer. BIOLOGY 2024; 13:716. [PMID: 39336143 PMCID: PMC11446462 DOI: 10.3390/biology13090716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
In recent years, knowledge of cell-released extracellular vesicle (EV) functions has undergone rapid growth. EVs are membrane vesicles loaded with proteins, nucleic acids, lipids, and bioactive molecules. Once released into the extracellular space, EVs are delivered to target cells that may go through modifications in physiological or pathological conditions. EVs are nano shuttles with a crucial role in promoting short- and long-distance cell-cell communication. Comprehension of the mechanism that regulates this process is a benefit for both medicine and basic science. Currently, EVs attract immense interest in precision and nanomedicine for their potential use in diagnosis, prognosis, and therapies. This review reports the latest advances in EV studies, focusing on the nature and features of EVs and on conventional and emerging methodologies used for their separation, characterization, and visualization. By searching an extended portion of the relevant literature, this work aims to give a summary of advances in nanomedical applications of EVs. Moreover, concerns that require further studies before translation to clinical applications are discussed.
Collapse
Affiliation(s)
- Maria Antonietta Di Bella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy
| | - Simona Taverna
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 90146 Palermo, Italy
| |
Collapse
|
41
|
Liu C, Zhou X, Zeng H, Yu J, Li W, Zhang W, Liao Y, Wang H, Liu L. Endoplasmic Reticulum Stress Potentiates the Immunosuppressive Microenvironment in Hepatocellular Carcinoma by Promoting the Release of SNHG6-Enriched Small Extracellular Vesicles. Cancer Immunol Res 2024; 12:1184-1201. [PMID: 38900485 DOI: 10.1158/2326-6066.cir-23-0469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 12/14/2023] [Accepted: 06/17/2024] [Indexed: 06/21/2024]
Abstract
Endoplasmic reticulum (ER) stress leads to hepatocellular carcinoma (HCC) progression. Small extracellular vesicles (sEV) play a crucial role in modulating the tumor microenvironment (TME) by influencing cellular communication and immune responses. However, it is unclear whether ER stress modulates the TME through sEVs. In the current study, we investigated the effects and underlying mechanisms of ER stress on the HCC TME. In vivo and in vitro experiments showed that overactivated ER stress was a salient attribute of the immunosuppressive HCC TME. This was caused by the ATF4-promoted release of small nucleolar RNA host gene 6 (SNHG6)-carrying sEVs, which attenuated T cell-mediated immune responses. Overall, SNHG6 modulated the immunosuppressive TME and aggravated ER stress. Meanwhile, targeting SNHG6 facilitated M1-like macrophage and CD8+ T-cell infiltration and decreased the proportion of M2-like macrophages. In addition, SNHG6 knockdown enhanced anti-PD1 immunotherapeutic efficacy. Moreover, in HCC patients, overexpression of SNHG6 was associated with a lack of response to anti-PD1 therapy and poor prognosis, whereas low SNHG6 expression was associated with improved therapeutic efficacy and prognoses. These data indicate that a correlation exists among ER stress, sEVs, immunosuppressive HCC TME, and immunotherapeutic efficacy. Hence, SNHG6-targeted therapy may represent an effective strategy for patients with HCC.
Collapse
Affiliation(s)
- Chengdong Liu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaohan Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Hanyi Zeng
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jiaping Yu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Wenwen Li
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Wanli Zhang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yanxia Liao
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Haijian Wang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Li Liu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
42
|
Huang S, Hu J, Hu M, Hou Y, Zhang B, Liu J, Liu X, Chen Z, Wang L. Cooperation between SIX1 and DHX9 transcriptionally regulates integrin-focal adhesion signaling mediated metastasis and sunitinib resistance in KIRC. Oncogene 2024; 43:2951-2969. [PMID: 39174859 DOI: 10.1038/s41388-024-03126-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/24/2024]
Abstract
High invasive capacity and acquired tyrosine kinase inhibitors (TKI) resistance of kidney renal clear cell carcinoma (KIRC) cells remain obstacles to prolonging the survival time of patients with advanced KIRC. In the present study, we reported that sine oculis homeobox 1 (SIX1) was upregulated in sunitinib-resistant KIRC cells and metastatic KIRC tissues. Subsequently, we found that SIX1 mediated metastasis and sunitinib resistance via Focal adhesion (FA) signaling, and knockdown of SIX1 enhanced the antitumor efficiency of sunitinib in KIRC. Mechanistically, Integrin subunit beta 1 (ITGB1), an upstream gene of FA signaling, was a direct transcriptional target of SIX1. In addition, we showed that DExH-box helicase 9 (DHX9) was an important mediator for SIX1-induced ITGB1 transcription, and silencing the subunits of SIX1/DHX9 complex significantly reduced transcription of ITGB1. Downregulation of SIX1 attenuated nuclear translocation of DHX9 and abrogated the binding of DHX9 to ITGB1 promoter. Collectively, our results unveiled a new signal axis SIX1/ITGB1/FAK in KIRC and identified a novel therapeutic strategy for metastatic KIRC patients.
Collapse
Affiliation(s)
- Shiyu Huang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, 430060, Wuhan, Hubei, China
| | - Juncheng Hu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Min Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yanguang Hou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Banghua Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, 430060, China
| | - Jiachen Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
43
|
Xie HJ, Jiang MJ, Jiang K, Tang LQ, Chen QY, Yang AK, Mai HQ. Communication between cancer cell subtypes by exosomes contributes to nasopharyngeal carcinoma metastasis and poor prognosis. PRECISION CLINICAL MEDICINE 2024; 7:pbae018. [PMID: 39347440 PMCID: PMC11427951 DOI: 10.1093/pcmedi/pbae018] [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: 05/05/2024] [Accepted: 08/25/2024] [Indexed: 10/01/2024] Open
Abstract
Background Intratumor heterogeneity is common in cancers, with different cell subtypes supporting each other to become more malignant. Nasopharyngeal carcinoma (NPC), a highly metastatic cancer, shows significant heterogeneity among its cells. This study investigates how NPC cell subtypes with varying metastatic potentials influence each other through exosome-transmitted molecules. Methods Exosomes were purified and characterized. MicroRNA expression was analyzed via sequencing and qRT-PCR. The effects of miR-30a-5p on migration, invasion, and metastasis were evaluated in vitro and in vivo. Its impact on desmoglein glycoprotein (DSG2) was assessed using dual-luciferase assays and Western blotting. Immunohistochemistry (IHC) and statistical models linked miR-30a-5p/DSG2 levels to patient prognosis. Results Different NPC cell subtypes transmit metastatic potential via exosomes. High-metastatic cells enhance the migration, invasion, and metastasis of low-metastatic cells through exosome-transmitted miR-30a-5p. Plasma levels of exosomal miR-30a-5p are reliable indicators of NPC prognosis. miR-30a-5p may promote metastasis by targeting DSG2 and modulating Wnt signaling. Plasma exosomal miR-30a-5p inversely correlates with DSG2 levels, predicting patient outcomes. Conclusion High-metastatic NPC cells can increase the metastatic potential of low-metastatic cells through exosome-transmitted miR-30a-5p, which is a valuable prognostic marker assessable via liquid biopsy.
Collapse
Affiliation(s)
- Hao-Jun Xie
- Department of Head and Neck, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ming-Jie Jiang
- Department of Head and Neck, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ke Jiang
- Department of Head and Neck, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Lin-Quan Tang
- Departcment of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Qiu-Yan Chen
- Departcment of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - An-Kui Yang
- Department of Head and Neck, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hai-Qiang Mai
- Departcment of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
44
|
González Á, López-Borrego S, Sandúa A, Vales-Gomez M, Alegre E. Extracellular vesicles in cancer: challenges and opportunities for clinical laboratories. Crit Rev Clin Lab Sci 2024; 61:435-457. [PMID: 38361287 DOI: 10.1080/10408363.2024.2309935] [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: 09/22/2023] [Revised: 01/03/2024] [Accepted: 01/22/2024] [Indexed: 02/17/2024]
Abstract
Extracellular vesicles (EVs) are nano-sized particles secreted by most cells. They transport different types of biomolecules (nucleic acids, proteins, and lipids) characteristic of their tissue or cellular origin that can mediate long-distance intercellular communication. In the case of cancer, EVs participate in tumor progression by modifying the tumor microenvironment, favoring immune tolerance and metastasis development. Consequently, EVs have great potential in liquid biopsy for cancer diagnosis, prognosis and follow-up. In addition, EVs could have a role in cancer treatment as a targeted drug delivery system. The intense research in the EV field has resulted in hundreds of patents and the creation of biomedical companies. However, methodological issues and heterogeneity in EV composition have hampered the advancement of EV validation trials and the development of EV-based diagnostic and therapeutic products. Consequently, only a few EV biomarkers have moved from research to clinical laboratories, such as the ExoDx Prostate IntelliScore (EPI) test, a CLIA/FDA-approved EV prostate cancer diagnostic test. In addition, the number of large-scale multicenter studies that would clearly define biomarker performance is limited. In this review, we will critically describe the different types of EVs, the methods for their enrichment and characterization, and their biological role in cancer. Then, we will specially focus on the parameters to be considered for the translation of EV biology to the clinic laboratory, the advances already made in the field of EVs related to cancer diagnosis and treatment, and the issues still pending to be solved before EVs could be used as a routine tool in oncology.
Collapse
Affiliation(s)
- Álvaro González
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Silvia López-Borrego
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council, Madrid, Spain
| | - Amaia Sandúa
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mar Vales-Gomez
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council, Madrid, Spain
| | - Estibaliz Alegre
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| |
Collapse
|
45
|
Hjazi A, Jasim SA, Altalbawy FMA, Kaur H, Hamzah HF, Kaur I, Deorari M, Kumar A, Elawady A, Fenjan MN. Relationship between lncRNA MALAT1 and Chemo-radiotherapy Resistance of Cancer Cells: Uncovered Truths. Cell Biochem Biophys 2024; 82:1613-1627. [PMID: 38806965 DOI: 10.1007/s12013-024-01317-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
The advancement of novel technologies, coupled with bioinformatics, has led to the discovery of additional genes, such as long noncoding RNAs (lncRNAs), that are associated with drug resistance. LncRNAs are composed of over 200 nucleotides and do not possess any protein coding function. These lncRNAs exhibit lower conservation across species, are typically expressed at low levels, and often display high specificity towards specific tissues and developmental stages. The LncRNA MALAT1 plays crucial regulatory roles in various aspects of genome function, encompassing gene transcription, splicing, and epigenetics. Additionally, it is involved in biological processes related to the cell cycle, cell differentiation, development, and pluripotency. Recently, MALAT1 has emerged as a novel mechanism contributing to drug resistance or sensitivity, attracting significant attention in the field of cancer research. This review aims to explore the mechanisms through which MALAT1 confers resistance to chemotherapy and radiotherapy in cancer cells.
Collapse
Affiliation(s)
- Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | | | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bangalore, Karnataka, India
- Faculty of Health and Life Sciences, Management and Science University, Shah Alam, Malaysia
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg, 620002, Russia
| | - Ahmed Elawady
- College of Technical Engineering, the Islamic University, Najaf, Iraq
- College of technical engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Mohammed N Fenjan
- College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| |
Collapse
|
46
|
Marima R, Basera A, Miya T, Damane BP, Kandhavelu J, Mirza S, Penny C, Dlamini Z. Exosomal long non-coding RNAs in cancer: Interplay, modulation, and therapeutic avenues. Noncoding RNA Res 2024; 9:887-900. [PMID: 38616862 PMCID: PMC11015109 DOI: 10.1016/j.ncrna.2024.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/20/2024] [Accepted: 03/29/2024] [Indexed: 04/16/2024] Open
Abstract
In the intricate field of cancer biology, researchers are increasingly intrigued by the emerging role of exosomal long non-coding RNAs (lncRNAs) due to their multifaceted interactions, complex modulation mechanisms, and potential therapeutic applications. These exosomal lncRNAs, carried within extracellular vesicles, play a vital partin tumorigenesis and disease progression by facilitating communication networks between tumor cells and their local microenvironment, making them an ideal candidates for use in a liquid biopsy approach. However, exosomal lncRNAs remain an understudied area, especially in cancer biology. Therefore this review aims to comprehensively explore the dynamic interplay between exosomal lncRNAs and various cellular components, including interactions with tumor-stroma, immune modulation, and drug resistance mechanisms. Understanding the regulatory functions of exosomal lncRNAs in these processes can potentially unveil novel diagnostic markers and therapeutic targets for cancer. Additionally, the emergence of RNA-based therapeutics presents exciting opportunities for targeting exosomal lncRNAs, offering innovative strategies to combat cancer progression and improve treatment outcomes. Thus, this review provides insights into the current understanding of exosomal lncRNAs in cancer biology, highlighting their crucial roles, regulatory mechanisms, and the evolving landscape of therapeutic interventions. Furthermore, we have also discussed the advantage of exosomes as therapeutic carriers of lncRNAs for the development of personalized targeted therapy for cancer patients.
Collapse
Affiliation(s)
- Rahaba Marima
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChi Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, South Africa
| | - Afra Basera
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChi Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, South Africa
- Department of Medical Oncology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, South Africa
| | - Thabiso Miya
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChi Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, South Africa
| | - Botle Precious Damane
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Pretoria, 0028, South Africa
| | - Jeyalakshmi Kandhavelu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Sheefa Mirza
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Parktown, 2193, South Africa
| | - Clement Penny
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Parktown, 2193, South Africa
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChi Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, South Africa
| |
Collapse
|
47
|
Ou J, Yin H, Shu F, Wu Z, Liu S, Ye J, Zhang S. Vasculogenic mimicry-related gene prognostic index for predicting prognosis, immune microenvironment in clear cell renal cell carcinoma. Heliyon 2024; 10:e36235. [PMID: 39247316 PMCID: PMC11380016 DOI: 10.1016/j.heliyon.2024.e36235] [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: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 09/10/2024] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is a highly aggressive cancer associated with higher death rates. However, traditional anti-angiogenic therapies have limited effectiveness due to drug resistance. Vascular mimicry (VM) provides a different way for tumors to develop blood vessels without relying on endothelial cells or angiogenesis. However, the intricate mechanisms and interplay between it and the immune microenvironment in ccRCC remain unclear. Methods A PubMed and GeneCards literature review was conducted to identify VM-related genes (VMRGs). VMRGs expression profiles were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), developing a novel VM risk score model and nomogram for ccRCC. The EBI ArrayExpress database (the validation set) was obtained to validate the prognostic model. The relationship between VMRGs risk score clinical characteristics and immune infiltration was investigated. Finally, the expression of six model VMRGs was validated using single-cell analysis, GEPIA, Human Protein Atlas (HPA), and quantitative Real-time PCR (qRT-PCR). Results Cox regression analysis and nomogram identified L1CAM, TEK, CLDN4, EFNA1, SERPINF1, and MALAT1 as independent prognostic risk factors, which could be used to stratify the ccRCC population into two risk groups with distinct immune profiles and responsiveness to immunotherapy. The results of single-cell analysis, GEPIA, HPA, and qRT-PCR validated the model genes' expression. Conclusions Our novel findings constructed a convenient and reliable 6 gene signatures as potential immunologic and prognostic biomarkers of VM in ccRCC.
Collapse
Affiliation(s)
- Junyong Ou
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Haoming Yin
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Fan Shu
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Zonglong Wu
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Shuai Liu
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Jianfei Ye
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Shudong Zhang
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| |
Collapse
|
48
|
Huang K, Yu L, Lu D, Zhu Z, Shu M, Ma Z. Long non-coding RNAs in ferroptosis, pyroptosis and necroptosis: from functions to clinical implications in cancer therapy. Front Oncol 2024; 14:1437698. [PMID: 39267831 PMCID: PMC11390357 DOI: 10.3389/fonc.2024.1437698] [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: 05/24/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
As global population ageing accelerates, cancer emerges as a predominant cause of mortality. Long non-coding RNAs (lncRNAs) play crucial roles in cancer cell growth and death, given their involvement in regulating downstream gene expression levels and numerous cellular processes. Cell death, especially non-apoptotic regulated cell death (RCD), such as ferroptosis, pyroptosis and necroptosis, significantly impacts cancer proliferation, invasion and metastasis. Understanding the interplay between lncRNAs and the diverse forms of cell death in cancer is imperative. Modulating lncRNA expression can regulate cancer onset and progression, offering promising therapeutic avenues. This review discusses the mechanisms by which lncRNAs modulate non-apoptotic RCDs in cancer, highlighting their potential as biomarkers for various cancer types. Elucidating the role of lncRNAs in cell death pathways provides valuable insights for personalised cancer interventions.
Collapse
Affiliation(s)
- Ke Huang
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Li Yu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Dingci Lu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Ziyi Zhu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Min Shu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Zhaowu Ma
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
49
|
Min L, Bu F, Meng J, Liu X, Guo Q, Zhao L, Li Z, Li X, Zhu S, Zhang S. Circulating small extracellular vesicle RNA profiling for the detection of T1a stage colorectal cancer and precancerous advanced adenoma. eLife 2024; 12:RP88675. [PMID: 39121006 PMCID: PMC11315448 DOI: 10.7554/elife.88675] [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] [Indexed: 08/11/2024] Open
Abstract
It takes more than 20 years for normal colorectal mucosa to develop into metastatic carcinoma. The long time window provides a golden opportunity for early detection to terminate the malignant progression. Here, we aim to enable liquid biopsy of T1a stage colorectal cancer (CRC) and precancerous advanced adenoma (AA) by profiling circulating small extracellular vesicle (sEV)-derived RNAs. We exhibited a full RNA landscape for the circulating sEVs isolated from 60 participants. A total of 58,333 annotated RNAs were detected from plasma sEVs, among which 1,615 and 888 sEV-RNAs were found differentially expressed in plasma from T1a stage CRC and AA compared to normal controls (NC). Then we further categorized these sEV-RNAs into six modules by a weighted gene coexpression network analysis and constructed a 60-gene t-SNE model consisting of the top 10 RNAs of each module that could well distinguish T1a stage CRC/AA from NC samples. Some sEV-RNAs were also identified as indicators of specific endoscopic and morphological features of different colorectal lesions. The top-ranked biomarkers were further verified by RT-qPCR, proving that these candidate sEV-RNAs successfully identified T1a stage CRC/AA from NC in another cohort of 124 participants. Finally, we adopted different algorithms to improve the performance of RT-qPCR-based models and successfully constructed an optimized classifier with 79.3% specificity and 99.0% sensitivity. In conclusion, circulating sEVs of T1a stage CRC and AA patients have distinct RNA profiles, which successfully enable the detection of both T1a stage CRC and AA via liquid biopsy.
Collapse
Affiliation(s)
- Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive DiseaseBeijingChina
- Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of SciencesBeijingChina
| | - Fanqin Bu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive DiseaseBeijingChina
| | - Jingxin Meng
- Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of SciencesBeijingChina
| | | | - Qingdong Guo
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive DiseaseBeijingChina
| | | | - Zhi Li
- Echo Biotech Co., LtdBeijingChina
| | - Xiangji Li
- Department of Retroperitoneal Tumor Surgery, International Hospital, Peking UniversityBeijingChina
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive DiseaseBeijingChina
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive DiseaseBeijingChina
| |
Collapse
|
50
|
Wang J, Dou P, Sun Y, Zheng J, Wu G, Liu H, Tao L. Epigenetic dysregulated long non-coding RNAs in renal cell carcinoma based on multi-omics data and their influence on target drugs sensibility. Front Genet 2024; 15:1406150. [PMID: 39156959 PMCID: PMC11327069 DOI: 10.3389/fgene.2024.1406150] [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: 03/24/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
Purpose Epigenetic modifications play a crucial role in cancer development, and our study utilized public data to analyze which leads to the discovery of significant epigenetic abnormalities in lncRNAs, offering valuable insights into prognosis and treatment strategies for renal carcinoma. Methods Public data were obtained from the Cancer Genome Atlas (TCGA), International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO) database. The analysis of the online public data was all completed in R software. Results We discovered a great number of epigenetic abnormalities of lncRNA in renal cancer, which is achieved by comparing the following modification and methylation of histone region changes on the promoter and enhancer of lncRNA: H3K27ac, H3K4me1, H3K4me3. As a result, 12 specific epigenetic disorders of lncRNA genes in renal cancer were identified. Finally, based on this lncRNA, we investigated the prognosis of renal cancer samples, among which 8 lncRNA can be seen as markers of prognosis in renal cancer, which had great prediction ability for ccRCC prognosis. Meanwhile, high risk score may pose response better to axitinib and nilotinib, but not sorafenib or sunitinib. Beyond, we observed an elevated level of risk score in immunotherapy non-responders. Further, biological enrichment and immuno-infiltration analysis was conducted to investigate the fundamental differences between patients categorized as high or low risk. Conclusion Our research improves the understanding in the function of epigenetic dysregulated long non-coding RNAs in renal carcinoma.
Collapse
Affiliation(s)
- Jiawei Wang
- Department of Urology, The Second People’s Hospital of Wuhu, Wuhu, China
| | - Pingnan Dou
- Department of Urology, The Second People’s Hospital of Wuhu, Wuhu, China
- School of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yunwen Sun
- Department of Urology, The Second People’s Hospital of Wuhu, Wuhu, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Jie Zheng
- Department of Urology, The Second People’s Hospital of Wuhu, Wuhu, China
| | - Guanwei Wu
- Department of Urology, The Second People’s Hospital of Wuhu, Wuhu, China
| | - Heqian Liu
- Department of Urology, The Second People’s Hospital of Wuhu, Wuhu, China
| | - Lingsong Tao
- Department of Urology, The Second People’s Hospital of Wuhu, Wuhu, China
| |
Collapse
|