1
|
Chen S, Wang Y, Zhang J, Liu B, Liu W, Cao G, Li R, Li H, Zhai N, Song X, Zhang S, Lv C. YTHDC1 phase separation drives the nuclear export of m 6A-modified lncNONMMUT062668.2 through the transport complex SRSF3-ALYREF-XPO5 to aggravate pulmonary fibrosis. Cell Death Dis 2025; 16:279. [PMID: 40221424 PMCID: PMC11993731 DOI: 10.1038/s41419-025-07608-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Fibroblast-to-myofibroblast differentiation is the main cytopathologic characteristic of pulmonary fibrosis. However, its underlying molecular mechanism remains poorly understood. This study elucidated that the nuclear export of lncNONMMUT062668.2 (lnc668) exacerbated pulmonary fibrosis by activating fibroblast-to-myofibroblast differentiation. Mechanistic research revealed that histone H3K9 lactylation in the promoter region of the N6-methyladenosine (m6A) writer METTL3 was enriched to enhance METTL3 transcription, leading to the lnc668 m6A modification. Meanwhile, the m6A reader YTHDC1 recognized m6A-modified lnc668 and elevated the METTL3-mediated lnc668 modification. Subsequently, phase-separating YTHDC1 promoted the nuclear export of m6A-modified lnc668. In this process, the phase-separating YTHDC1 formed a nuclear pore complex with serine/arginine-rich splicing factor 3, Aly/REF export factor, and exportin-5 to assist the translocation of m6A-modified lnc668 from nucleus to cytoplasm. After nuclear export, lnc668 facilitated the translation and stability of its host gene phosphatidylinositol-binding clathrin assembly protein to activate fibroblast-to-myofibroblast differentiation, leading to the aggravation of pulmonary fibrosis, which also depended on YTHDC1 phase separation. This study first clarified that YTHDC1 phase separation is crucial for the m6A modification, nuclear export, and profibrotic role of lnc668 in exacerbating pulmonary fibrosis. These findings provide new insights into the nuclear export of cytoplasmic lncRNAs and identified potential targets for pulmonary fibrosis therapy.
Collapse
Affiliation(s)
- Shengjun Chen
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Yujie Wang
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai, China
| | - Jinjin Zhang
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai, China
| | - Bo Liu
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Weili Liu
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Guohong Cao
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Rongrong Li
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Hongbo Li
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Nailiang Zhai
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Xiaodong Song
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai, China.
| | - Songzi Zhang
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai, China.
- CHA Bundang Medical Center, CHA University, Seongnam-si, Republic of Korea.
| | - Changjun Lv
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China.
| |
Collapse
|
2
|
Quaresima B, Scicchitano S, Faniello MC, Mesuraca M. Role of solute carrier transporters in ovarian cancer (Review). Int J Mol Med 2025; 55:24. [PMID: 39611477 PMCID: PMC11637498 DOI: 10.3892/ijmm.2024.5465] [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: 08/23/2024] [Accepted: 10/11/2024] [Indexed: 11/30/2024] Open
Abstract
Solute carrier (SLC) transporters are involved in various biological processes associated with metabolic reprogramming and cancer, supporting the increased requirement of nutrients and energy. Over the past decade, there have been significant advancements in understanding the expression and function of SLCs in ovarian cancer (OC). This gynecological condition has a high mortality rate and limited treatment options; thus, early diagnosis remains a target clinically. OC exhibits complexity and heterogeneity, resulting in different clinical characteristics, resistance to chemotherapy drugs and poor prognosis. Additionally, SLCs have a different expression pattern between healthy and tumor tissue, and consequently, their inhibition or activation could modify signaling pathways involved in the tumor growth process, such as cell proliferation, apoptosis and drug accumulation. The present review aims to consolidate current data to provide a comprehensive understanding of the potential importance of SLCs in OC. Additionally, it seeks to offer guidance for further research on utilizing SLCs as prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Barbara Quaresima
- Department of Experimental and Clinical Medicine, 'Magna Graecia' University of Catanzaro, I-88100 Catanzaro, Italy
| | - Stefania Scicchitano
- Department of Experimental and Clinical Medicine, 'Magna Graecia' University of Catanzaro, I-88100 Catanzaro, Italy
| | | | - Maria Mesuraca
- Correspondence to: Dr Maria Mesuraca or Dr Barbara Quaresima, Department of Experimental and Clinical Medicine, 'Magna Graecia' University of Catanzaro, Viale Europa, I-88100 Catanzaro, Italy, E-mail: , E-mail:
| |
Collapse
|
3
|
Ni Y, Wang W, Jiang L, Shao Q. Exploring the molecular interface of gene expression dynamics and prostate cancer susceptibility in response to HBCD exposure. Toxicol Res (Camb) 2025; 14:tfaf016. [PMID: 39906184 PMCID: PMC11788417 DOI: 10.1093/toxres/tfaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/15/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025] Open
Abstract
Hexabromocyclododecane (HBCD), a brominated flame retardant, is linked to various health implications, including prostate cancer. This study explored the molecular mechanisms and potential biomarkers associated with HBCD exposure using data from the Comparative Toxicogenomics Database (CTD) and The Cancer Genome Atlas (TCGA). A total of 7,147 differentially expressed genes (DEGs) and 46 differentially expressed miRNAs were identified, with significant enrichment in cancer-related pathways and xenobiotic metabolism. Protein-protein interaction (PPI) network construction and enrichment analyses revealed four hub genes: DNAJC12, PKMYT1, RRM2, and SLC12A5. These genes displayed notable expression changes in response to HBCD exposure and were strongly correlated with survival outcomes in prostate cancer patients, as demonstrated by Cox regression and ROC curve analyses. Additionally, miRNA correlation analyses indicated robust positive associations, highlighting a coordinated regulatory network. Experimental expression analyses on HBCD-treated cell lines further validated these findings. This study sheds light on the significant impact of HBCD on gene and miRNA expression in prostate cancer, emphasizing the potential of the identified hub genes and miRNAs as prognostic biomarkers and therapeutic targets. By elucidating the pathways and regulatory networks influenced by HBCD, the findings provide a foundation for developing strategies to mitigate its carcinogenic effects and improve outcomes for prostate cancer patients.
Collapse
Affiliation(s)
- Ying Ni
- Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing 100875, China
| | - Wenkai Wang
- Department of Oncology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Pudong New Area, Shanghai 201203, China
| | - Lihua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Cross Street, Hongshan Road, Jiangsu Province, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Cross Street, Hongshan Road, Jiangsu Province, Nanjing 210028, China
| | - Qinghua Shao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Cross Street, Hongshan Road, Jiangsu Province, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Cross Street, Hongshan Road, Jiangsu Province, Nanjing 210028, China
| |
Collapse
|
4
|
Uddin MB, Wang Z, Yang C. Epitranscriptomic RNA m 6A Modification in Cancer Therapy Resistance: Challenges and Unrealized Opportunities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 12:e2403936. [PMID: 39661414 PMCID: PMC11775542 DOI: 10.1002/advs.202403936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/24/2024] [Indexed: 12/12/2024]
Abstract
Significant advances in the development of new cancer therapies have given rise to multiple novel therapeutic options in chemotherapy, radiotherapy, immunotherapy, and targeted therapies. Although the development of resistance is often reported along with temporary disease remission, there is often tumor recurrence of an even more aggressive nature. Resistance to currently available anticancer drugs results in poor overall and disease-free survival rates for cancer patients. There are multiple mechanisms through which tumor cells develop resistance to therapeutic agents. To date, efforts to overcome resistance have only achieved limited success. Epitranscriptomics, especially related to m6A RNA modification dysregulation in cancer, is an emerging mechanism for cancer therapy resistance. Here, recent studies regarding the contributions of m6A modification and its regulatory proteins to the development of resistance to different cancer therapies are comprehensively reviewed. The promise and potential limitations of targeting these entities to overcome resistance to various anticancer therapies are also discussed.
Collapse
Affiliation(s)
- Mohammad Burhan Uddin
- Department of Pharmaceutical SciencesNorth South UniversityBashundharaDhaka1229Bangladesh
| | - Zhishan Wang
- Stony Brook Cancer CenterStony Brook UniversityStony BrookNY11794USA
| | - Chengfeng Yang
- Stony Brook Cancer CenterStony Brook UniversityStony BrookNY11794USA
- Department of PathologyRenaissance School of MedicineStony Brook UniversityStony BrookNY11794USA
| |
Collapse
|
5
|
Wang H, Dai Y, Wang F. ETV4‑mediated transcriptional activation of SLC12A5 exacerbates ferroptosis resistance and glucose metabolism reprogramming in breast cancer cells. Mol Med Rep 2024; 30:217. [PMID: 39370816 PMCID: PMC11465427 DOI: 10.3892/mmr.2024.13341] [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: 01/18/2024] [Accepted: 05/20/2024] [Indexed: 10/08/2024] Open
Abstract
Solute carrier family 12 member 5 (SLC12A5) is an oncogene in numerous types of cancer, however its function in breast cancer (BC) remains elusive. ETS translocation variant 4 (ETV4) promotes BC. Therefore, the present study aimed to elucidate the role of SLC12A5 in ferroptosis and glucose metabolism in BC cells as well as to understand the underlying mechanism. Analysis of data from the UALCAN database demonstrated expression levels of SLC12A5 in BC and its association with prognosis. Reverse transcription‑quantitative PCR and western blotting were conducted to evaluate the expression levels of SLC12A5 and ETV4 in BC cells. The abilities of BC cells to proliferate, migrate and invade were assessed using Cell Counting Kit‑8, colony formation, wound healing and Transwell assays. Thiobarbituric acid reactive substances assay and a C11 BODIPY 581/591 probe were used to evaluate lipid peroxidation. Ferroptosis resistance was evaluated by the measurement of Fe2+ and ferroptosis‑related solute carrier family 7a member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), acyl‑CoA synthetase long‑chain family member 4 (ACSL4) and transferrin receptor 1 (TFR1) protein levels. Glycolysis was assessed via evaluation of extracellular acidification rate, oxygen consumption rate, lactate production and glucose consumption. Finally, luciferase reporter and chromatin immunoprecipitation assay were used to verify the interaction between ETV4 and the SLC12A5 promoter. UALCAN database analysis indicated that SLC12A5 was upregulated in BC tissues and cells and that SLC12A5 elevation indicated a poor prognosis of patients with BC. SLC12A5 knockdown suppressed the BC cell proliferative, migratory and invasive capabilities. Moreover, SLC12A5 knockdown decreased BC cell ferroptosis resistance and glucose metabolism reprogramming. The transcription factor ETV4 was demonstrated to bind to the SLC12A5 promoter and upregulate its transcription. Furthermore, ETV4 overexpression counteracted the suppressive effect of SLC12A5 knockdown on the BC cell proliferative, migratory and invasive abilities, as well as on ferroptosis resistance and glucose metabolism reprogramming. Transcriptional activation of SLC12A5 by ETV4 modulated the migration, invasion, ferroptosis resistance and glucose metabolism reprogramming of BC cells.
Collapse
Affiliation(s)
- Huan Wang
- Department of Pathology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Yanyan Dai
- Department of Pathology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Fengxiang Wang
- Department of Pathology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
6
|
Sharma S. Unraveling the role of long non-coding RNAs in therapeutic resistance in acute myeloid leukemia: New prospects & challenges. Noncoding RNA Res 2024; 9:1203-1221. [PMID: 39036603 PMCID: PMC11259994 DOI: 10.1016/j.ncrna.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 07/23/2024] Open
Abstract
Acute Myeloid Leukemia (AML) is a fatal hematological disease characterized by the unchecked proliferation of immature myeloid blasts in different tissues developed by various mutations in hematopoiesis. Despite intense chemotherapeutic regimens, patients often experience poor outcomes, leading to substandard remission rates. In recent years, long non-coding RNAs (lncRNAs) have increasingly become important prognostic and therapeutic hotspots, due to their contributions to dysregulating many functional epigenetic, transcriptional, and post-translational mechanisms leading to alterations in cell expressions, resulting in increased chemoresistance and reduced apoptosis in leukemic cells. Through this review, I highlight and discuss the latest advances in understanding the major mechanisms through which lncRNAs confer therapy resistance in AML. In addition, I also provide perspective on the current strategies to target lncRNA expressions. A better knowledge of the critical role that lncRNAs play in controlling treatment outcomes in AML will help improve existing medications and devise new ones.
Collapse
Affiliation(s)
- Siddhant Sharma
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| |
Collapse
|
7
|
Chen D, Gu X, Nurzat Y, Xu L, Li X, Wu L, Jiao H, Gao P, Zhu X, Yan D, Li S, Xue C. Writers, readers, and erasers RNA modifications and drug resistance in cancer. Mol Cancer 2024; 23:178. [PMID: 39215288 PMCID: PMC11363509 DOI: 10.1186/s12943-024-02089-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Drug resistance in cancer cells significantly diminishes treatment efficacy, leading to recurrence and metastasis. A critical factor contributing to this resistance is the epigenetic alteration of gene expression via RNA modifications, such as N6-methyladenosine (m6A), N1-methyladenosine (m1A), 5-methylcytosine (m5C), 7-methylguanosine (m7G), pseudouridine (Ψ), and adenosine-to-inosine (A-to-I) editing. These modifications are pivotal in regulating RNA splicing, translation, transport, degradation, and stability. Governed by "writers," "readers," and "erasers," RNA modifications impact numerous biological processes and cancer progression, including cell proliferation, stemness, autophagy, invasion, and apoptosis. Aberrant RNA modifications can lead to drug resistance and adverse outcomes in various cancers. Thus, targeting RNA modification regulators offers a promising strategy for overcoming drug resistance and enhancing treatment efficacy. This review consolidates recent research on the role of prevalent RNA modifications in cancer drug resistance, with a focus on m6A, m1A, m5C, m7G, Ψ, and A-to-I editing. Additionally, it examines the regulatory mechanisms of RNA modifications linked to drug resistance in cancer and underscores the existing limitations in this field.
Collapse
Affiliation(s)
- Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yeltai Nurzat
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lixia Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xueyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Lixin Wu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Henan Jiao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Peng Gao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xuqiang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Shaohua Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
8
|
Cao Y, Jia M, Duan C, Yang Z, Cheng B, Wang R. The m 6A regulators in prostate cancer: molecular basis and clinical perspective. Front Pharmacol 2024; 15:1448872. [PMID: 39268470 PMCID: PMC11391310 DOI: 10.3389/fphar.2024.1448872] [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: 06/14/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death among men in western countries. Evidence has indicated the significant role of the androgen receptor (AR) as the main driving factor in controlling the development of PCa, making androgen receptor inhibition (ARI) therapy a pivotal management approach. In addition, AR independent signaling pathways also contribute to PCa progression. One such signaling pathway that has garnered our attention is N6-Methyladenosine (m6A) signaling, which refers to a chemical modification on RNA with crucial roles in RNA metabolism and disease progression, including PCa. It is important to comprehensively summarize the role of each individual m6A regulator in PCa development and understand its interaction with AR signaling. This review aims to provide a thorough summary of the involvement of m6A regulators in PCa development, shedding light on their upstream and downstream signaling pathways. This summary sets the stage for a comprehensive review that would benefit the scientific community and clinical practice by enhancing our understanding of the biology of m6A regulators in the context of PCa.
Collapse
Affiliation(s)
- Yu Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Man Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Chunyan Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Zhihui Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Cheng
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ronghao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
9
|
Yang L, Ying J, Tao Q, Zhang Q. RNA N 6-methyladenosine modifications in urological cancers: from mechanism to application. Nat Rev Urol 2024; 21:460-476. [PMID: 38347160 DOI: 10.1038/s41585-023-00851-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 08/04/2024]
Abstract
The N6-methyladenosine (m6A) modification is the most common modification of messenger RNAs in eukaryotes and has crucial roles in multiple cancers, including in urological malignancies such as renal cell carcinoma, bladder cancer and prostate cancer. The m6A RNA modification is controlled by three types of regulators, including methyltransferases (writers), demethylases (erasers) and RNA-binding proteins (readers), which are responsible for gene regulation at the post-transcriptional level. This Review summarizes the current evidence indicating that aberrant or dysregulated m6A modification is associated with urological cancer development, progression and prognosis. The complex and context-dependent effects of dysregulated m6A modifications in urological cancers are described, along with the potential for aberrantly expressed m6A regulators to provide valuable diagnostic and prognostic biomarkers as well as new therapeutic targets.
Collapse
Affiliation(s)
- Lei Yang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Jianming Ying
- Department of Pathology, Cancer Institute and Cancer Hospital, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Tao
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Qian Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China.
- Department of Urology, Peking University Binhai Hospital, Tianjin, China.
| |
Collapse
|
10
|
Wu B, Dou X, Zhao Y, Wang X, Zhao C, Xia J, Xing C, He S, Feng C. Chiral Supramolecular Nanofibers Regulated Tumor-Derived Exosomes Secretion for Constructing an Anti-Tumor Extracellular Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308335. [PMID: 38420895 DOI: 10.1002/smll.202308335] [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: 09/21/2023] [Revised: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Tumor-derived exosomes (TDEs) induced extracellular microenvironment has recently been validated to be critical for tumor progression and metastasis, however, remodeling it for oncotherapy still remains a major challenge due to difficulty in regulation of TDEs secretion. Herein, the supramolecular chiral nanofibers, composed of L/D-phenylalanine derivates (L/D-Phe) and linear hyaluronic acid (HA), are successfully employed to construct TDEs induced anti-tumor extracellular microenvironment. The left-handed L-Phe @HA nanofibers significantly inhibit TDEs secretion into extracellular microenvironment, which results in suppression of tumor proliferation and metastasis in vitro and vivo. Biological assays and theoretical modeling reveal that these results are mainly attributed to strong adsorption of the key exosomes transporters (Ras-related protein Rab-27A and synaptosome-associated protein 23) on left-handed L-Phe @HA nanofibers via enhanced stereoselective interaction, leading to degradation and phosphorylated dropping of exosomes transporters. Subsequently, transfer function of exosomes transporters is limited, which causes remarkable inhibition of TDEs secretion. These findings provide a promising novel insight of chiral functional materials to establish an anti-tumor extracellular microenvironment via regulation of TDEs secretion.
Collapse
Affiliation(s)
- Beibei Wu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Xiaoqiu Dou
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Yu Zhao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Xueqian Wang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Changli Zhao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Jingyi Xia
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Chao Xing
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Sijia He
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Chuanliang Feng
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| |
Collapse
|
11
|
Zhang H, Wang Z, Qiao X, Peng N, Wu J, Chen Y, Cheng C. Unveiling the therapeutic potential of IHMT-337 in glioma treatment: targeting the EZH2-SLC12A5 axis. Mol Med 2024; 30:91. [PMID: 38886655 PMCID: PMC11184773 DOI: 10.1186/s10020-024-00857-0] [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: 04/28/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
Glioma is the most common malignant tumor of the central nervous system, with EZH2 playing a crucial regulatory role. This study further explores the abnormal expression of EZH2 and its mechanisms in regulating glioma progression. Additionally, it was found that IHMT-337 can potentially be a therapeutic agent for glioma. The prognosis, expression, and localization of EZH2 were determined using bioinformatics, IHC staining, Western blot (WB) analysis, and immunofluorescence (IF) localization. The effects of EZH2 on cell function were assessed using CCK-8 assays, Transwell assays, and wound healing assays. Public databases and RT-qPCR were utilized to identify downstream targets. The mechanisms regulating these downstream targets were elucidated using MS-PCR and WB analysis. The efficacy of IHMT-337 was demonstrated through IC50 measurements, WB analysis, and RT-qPCR. The effects of IHMT-337 on glioma cells in vitro were evaluated using Transwell assays, EdU incorporation assays, and flow cytometry. The potential of IHMT-337 as a treatment for glioma was assessed using a blood-brain barrier (BBB) model and an orthotopic glioma model. Our research confirms significantly elevated EZH2 expression in gliomas, correlating with patient prognosis. EZH2 facilitates glioma proliferation, migration, and invasion alongside promoting SLC12A5 DNA methylation. By regulating SLC12A5 expression, EZH2 activates the WNK1-OSR1-NKCC1 pathway, enhancing its interaction with ERM to promote glioma migration. IHMT-337 targets EZH2 in vitro to inhibit WNK1 activation, thereby suppressing glioma cell migration. Additionally, it inhibits cell proliferation and arrests the cell cycle. IHMT-337 has the potential to cross the BBB and has successfully inhibited glioma progression in vivo. This study expands our understanding of the EZH2-SLC12A5 axis in gliomas, laying a new foundation for the clinical translation of IHMT-337 and offering new insights for precision glioma therapy.
Collapse
Affiliation(s)
- Hongwei Zhang
- Anhui University of Science and Technology, Huainan, 232001, Anhui, China
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Zixuan Wang
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Xiaolong Qiao
- Anhui University of Science and Technology, Huainan, 232001, Anhui, China
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Nan Peng
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Jiaxing Wu
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Yinan Chen
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Chuandong Cheng
- Anhui University of Science and Technology, Huainan, 232001, Anhui, China.
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
12
|
Pan J, Tong F, Ren N, Ren L, Yang Y, Gao F, Xu Q. Role of N 6‑methyladenosine in the pathogenesis, diagnosis and treatment of prostate cancer (Review). Oncol Rep 2024; 51:88. [PMID: 38757383 PMCID: PMC11110010 DOI: 10.3892/or.2024.8747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Prostate cancer (PCa) affects males of all racial and ethnic groups, and leads to higher rates of mortality in those belonging to a lower socioeconomic status due to the late detection of the disease. PCa affects middle‑aged males between the ages of 45 and 60 years, and is the highest cause of cancer‑associated mortality in Western countries. As the most abundant and common mRNA modification in higher eukaryotes, N6‑methyladenosine (m6A) is widely distributed in mammalian cells and influences various aspects of mRNA metabolism. Recent studies have found that abnormal expression levels of various m6A regulators significantly affect the development and progression of various types of cancer, including PCa. The present review discusses the influence of m6A regulatory factors on the pathogenesis and progression of PCa through mRNA modification based on the current state of research on m6A methylation modification in PCa. It is considered that the treatment of PCa with micro‑molecular drugs that target the epigenetics of the m6A regulator to correct abnormal m6A modifications is a direction for future research into current diagnostic and therapeutic approaches for PCa.
Collapse
Affiliation(s)
- Junjie Pan
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310051, P.R. China
| | - Fei Tong
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Ning Ren
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310051, P.R. China
| | - Lanqi Ren
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310051, P.R. China
| | - Yibei Yang
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310051, P.R. China
| | - Feng Gao
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, P.R. China
| | - Qiaoping Xu
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
13
|
Zhang Z, Liu J, Wu Y, Gu Z, Zou L, Liu Y, Geng J, Mao S, Luo M, Guo C, Zhang W, Yao X. The functions and mechanisms of RNA modification in prostate: Current status and future perspectives. Front Genet 2024; 15:1380746. [PMID: 38798700 PMCID: PMC11116725 DOI: 10.3389/fgene.2024.1380746] [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: 02/02/2024] [Accepted: 04/11/2024] [Indexed: 05/29/2024] Open
Abstract
The increasing incidence and mortality of prostate cancer worldwide significantly impact the life span of male patients, emphasizing the urgency of understanding its pathogenic mechanism and associated molecular changes that regulate tumor progression for effective prevention and treatment. RNA modification, an important post-transcriptional regulatory process, profoundly influences tumor cell growth and metabolism, shaping cell fate. Over 170 RNA modification methods are known, with prominent research focusing on N6-methyladenosine, N7-methylguanosine, N1-methyladenosine, 5-methylcytidine, pseudouridine, and N4-acetylcytidine modifications. These alterations intricately regulate coding and non-coding RNA post-transcriptionally, affecting the stability of RNA and protein expression levels. This article delves into the latest advancements and challenges associated with various RNA modifications in prostate cancer tumor cells, tumor microenvironment, and core signaling molecule androgen receptors. It aims to provide new research targets and avenues for molecular diagnosis, treatment strategies, and improvement of the prognosis in prostate cancer.
Collapse
Affiliation(s)
- Zhijin Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Ji Liu
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Yang Wu
- School of Medicine, Tongji University, Shanghai, China
| | - Zhuoran Gu
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Libin Zou
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Yingdi Liu
- Department of Pathology, Shanghai Tenth People’s Hospital, Shanghai, China
| | - Jiang Geng
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Shiyu Mao
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Ming Luo
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Changcheng Guo
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Urologic Cancer Institute, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
14
|
Liu H, Pan Z, Lin X, Chen L, Yang Q, Zhang W, Dai L, Zhang Y, Li W, Chen Y, Peng K, Wanggou S, Zeng F, Li X. A potassium-chloride co-transporter with altered genome architecture functions as a suppressor in glioma. J Cell Mol Med 2024; 28:e18352. [PMID: 38685685 PMCID: PMC11058328 DOI: 10.1111/jcmm.18352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/28/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
Gliomas, the most lethal tumours in brain, have a poor prognosis despite accepting standard treatment. Limited benefits from current therapies can be attributed to genetic, epigenetic and microenvironmental cues that affect cell programming and drive tumour heterogeneity. Through the analysis of Hi-C data, we identified a potassium-chloride co-transporter SLC12A5 associated with disrupted topologically associating domain which was downregulated in tumour tissues. Multiple independent glioma cohorts were included to analyse the characterization of SLC12A5 and found it was significantly associated with pathological features, prognostic value, genomic alterations, transcriptional landscape and drug response. We constructed two SLC12A5 overexpression cell lines to verify the function of SLC12A5 that suppressed tumour cell proliferation and migration in vitro. In addition, SLC12A5 was also positively associated with GABAA receptor activity and negatively associated with pro-tumour immune signatures and immunotherapy response. Collectively, our study provides a comprehensive characterization of SLC12A5 in glioma and supports SLC12A5 as a potential suppressor of disease progression.
Collapse
Affiliation(s)
- Hongwei Liu
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Zhouyang Pan
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Xuelei Lin
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Long Chen
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Qi Yang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Wei Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Luohuan Dai
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Yihao Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Wang Li
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Yinhua Chen
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Kang Peng
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
- Department of Radiology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Siyi Wanggou
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| | - Feiyue Zeng
- Department of Radiology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Xuejun Li
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
15
|
Hacioglu C, Oral D. Borax affects cellular viability by inducing ER stress in hepatocellular carcinoma cells by targeting SLC12A5. J Cell Mol Med 2024; 28:e18380. [PMID: 38780503 PMCID: PMC11114215 DOI: 10.1111/jcmm.18380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 04/22/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024] Open
Abstract
Hepatocellular carcinoma (HCC) presents a persistent challenge to conventional therapeutic approaches. SLC12A5 is implicated in an oncogenic capacity and facilitates the progression of cancer. The objective of this investigation is to scrutinize the inhibitory effects of borax on endoplasmic reticulum (ER)-stress and apoptosis mediated by SLC12A5 in HepG2 cells. Initially, we evaluated the cytotoxic impact of borax on both HL-7702 and HepG2 cell lines. Subsequently, the effects of borax on cellular morphology and the cell cycle of these lines were examined. Following this, we explored the impact of borax treatment on the mRNA and protein expression levels of SLC12A5, C/EBP homologous protein (CHOP), glucose-regulated protein-78 (GRP78), activating transcription factor-6 (ATF6), caspase-3 (CASP3), and cytochrome c (CYC) in these cellular populations. The determined IC50 value of borax for HL-7702 cells was 40.8 mM, whereas for HepG2 cells, this value was 22.6 mM. The concentrations of IC50 (22.6 mM) and IC75 (45.7 mM) of borax in HepG2 cells did not manifest morphological aberrations in HL-7702 cells. Conversely, these concentrations in HepG2 cells induced observable morphological and nuclear abnormalities, resulting in cell cycle arrest in the G1/G0 phase. Additionally, the levels of SLC12A5, ATF6, CHOP, GRP78, CASP3, and CYC were elevated in HepG2 cells in comparison to HL-7702 cells. Moreover, SLC12A5 levels decreased following borax treatment in HepG2 cells, whereas ATF6, CHOP, GRP78, CASP3, and CYC levels exhibited a significant increase. In conclusion, our data highlight the potential therapeutic effects of borax through the regulation of ER stress in HCC by targeting SLC12A5.
Collapse
Affiliation(s)
- Ceyhan Hacioglu
- Faculty of Pharmacy, Department of BiochemistryDüzce UniversityDüzceTurkey
- Faculty of Medicine, Department of Medical BiochemistryDüzce UniversityDüzceTurkey
| | - Didem Oral
- Faculty of Pharmacy, Department of Pharmaceutical ToxicologyDüzce UniversityDüzceTurkey
| |
Collapse
|
16
|
Li R, Zhu C, Wang Y, Wang X, Wang Y, Wang J, Wang K. The relationship between the network of non-coding RNAs-molecular targets and N6-methyladenosine modification in tumors of urinary system. Cell Death Dis 2024; 15:275. [PMID: 38632251 PMCID: PMC11024199 DOI: 10.1038/s41419-024-06664-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: 11/02/2023] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
N6-methyladenosine (m6A) methylation, a prevalent eukaryotic post-transcriptional modification, is involved in multiple biological functions, including mediating variable splicing, RNA maturation, transcription, and nuclear export, and also is vital for regulating RNA translation, stability, and cytoplasmic degradation. For example, m6A methylation can regulate pre-miRNA expression by affecting both splicing and maturation. Non-coding RNA (ncRNA), which includes microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), does not encode proteins but has powerful impacts on transcription and translation. Conversely, ncRNAs may impact m6A methylation by affecting the expression of m6A regulators, including miRNAs targeting mRNA of m6A regulators, or lncRNAs, and circRNAs, acting as scaffolds to regulate transcription of m6A regulatory factors. Dysregulation of m6A methylation is common in urinary tumors, and the regulatory role of ncRNAs is also important for these malignancies. This article provides a systematic review of the role and mechanisms of action of m6A methylation and ncRNAs in urinary tumors.
Collapse
Affiliation(s)
- Ruiming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Chunming Zhu
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yuan Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xia Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yibing Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Jiahe Wang
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
17
|
Ashrafizadeh M, Zhang W, Tian Y, Sethi G, Zhang X, Qiu A. Molecular panorama of therapy resistance in prostate cancer: a pre-clinical and bioinformatics analysis for clinical translation. Cancer Metastasis Rev 2024; 43:229-260. [PMID: 38374496 DOI: 10.1007/s10555-024-10168-9] [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: 06/09/2023] [Accepted: 01/04/2024] [Indexed: 02/21/2024]
Abstract
Prostate cancer (PCa) is a malignant disorder of prostate gland being asymptomatic in early stages and high metastatic potential in advanced stages. The chemotherapy and surgical resection have provided favourable prognosis of PCa patients, but advanced and aggressive forms of PCa including CRPC and AVPC lack response to therapy properly, and therefore, prognosis of patients is deteriorated. At the advanced stages, PCa cells do not respond to chemotherapy and radiotherapy in a satisfactory level, and therefore, therapy resistance is emerged. Molecular profile analysis of PCa cells reveals the apoptosis suppression, pro-survival autophagy induction, and EMT induction as factors in escalating malignant of cancer cells and development of therapy resistance. The dysregulation in molecular profile of PCa including upregulation of STAT3 and PI3K/Akt, downregulation of STAT3, and aberrant expression of non-coding RNAs are determining factor for response of cancer cells to chemotherapy. Because of prevalence of drug resistance in PCa, combination therapy including co-utilization of anti-cancer drugs and nanotherapeutic approaches has been suggested in PCa therapy. As a result of increase in DNA damage repair, PCa cells induce radioresistance and RelB overexpression prevents irradiation-mediated cell death. Similar to chemotherapy, nanomaterials are promising for promoting radiosensitivity through delivery of cargo, improving accumulation in PCa cells, and targeting survival-related pathways. In respect to emergence of immunotherapy as a new tool in PCa suppression, tumour cells are able to increase PD-L1 expression and inactivate NK cells in mediating immune evasion. The bioinformatics analysis for evaluation of drug resistance-related genes has been performed.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Yu Tian
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Xianbin Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China.
| | - Aiming Qiu
- Department of Geriatrics, the Fifth People's Hospital of Wujiang District, Suzhou, China.
| |
Collapse
|
18
|
Han Z, Yi X, Li J, Zhang T, Liao D, You J, Ai J. RNA m 6A modification in prostate cancer: A new weapon for its diagnosis and therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:188961. [PMID: 37507057 DOI: 10.1016/j.bbcan.2023.188961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/21/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
Prostate cancer (PCa) is the most common malignant tumor and the second leading cause of cancer-related mortality in men worldwide. Despite significant advances in PCa therapy, the underlying molecular mechanisms have yet to be fully elucidated. Recently, epigenetic modification has emerged as a key player in tumor progression, and RNA-based N6-methyladenosine (m6A) epigenetic modification was found to be crucial. This review summarizes comprehensive state-of-art mechanisms underlying m6A modification, its implication in the pathogenesis, and advancement of PCa in protein-coding and non-coding RNA contexts, its relevance to PCa immunotherapy, and the ongoing clinical trials for PCa treatment. This review presents potential m6A-based targets and paves a new avenue for diagnosing and treating PCa, providing new guidelines for future related research through a systematic review of previous results.
Collapse
Affiliation(s)
- Zeyu Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Xianyanling Yi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jin Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Tianyi Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Dazhou Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jia You
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China.
| |
Collapse
|
19
|
Zhu W, Zhao R, Guan X, Wang X. The emerging roles and mechanism of N6-methyladenosine (m 6A) modifications in urologic tumours progression. Front Pharmacol 2023; 14:1192495. [PMID: 37284313 PMCID: PMC10239868 DOI: 10.3389/fphar.2023.1192495] [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: 03/23/2023] [Accepted: 05/09/2023] [Indexed: 06/08/2023] Open
Abstract
Prostate cancer (PCa), bladder cancer (BC), and renal cell cancer (RCC) are the most common urologic tumours in males. N6-methyladenosine (m6A), adenosine N6 methylation, is the most prevalent RNA modification in mammals. Increasing evidence suggests that m6A plays a crucial role in cancer development. In this review, we comprehensively analyzed the influence of m6A methylation on Prostate cancer, bladder cancer, and renal cell cancer and the relationship between the expression of relevant regulatory factors and their development and occurrence, which provides new insights and approaches for the early clinical diagnosis and targeted therapy of urologic malignancies.
Collapse
|