1
|
Zhang F, Zhu T, Gan L, Du Y, Liu L, Ding H, Chen X, Guan Y. FTO triggers NLRP3/GSDMD-dependent pyroptosis to enhance cisplatin-sensitivity in ovarian cancer. Cell Signal 2025; 131:111698. [PMID: 39999914 DOI: 10.1016/j.cellsig.2025.111698] [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: 04/23/2024] [Revised: 02/15/2025] [Accepted: 02/20/2025] [Indexed: 02/27/2025]
Abstract
Chemotherapy resistance is still the major impediment to poor prognosis of ovarian cancer (OC). The fat mass and obesity associated protein (FTO), as the first recognized RNA N6-methyladenosine (m6A) demethylase, was dysregulated in various tumors, but its effect on chemoresistance of OC remains unexplored. In this study, FTO expression in tissues and cells was measured by qPCR, IHC, and Western blot. Functional assays were performed, including CCK-8, colony formation, flow cytometry, Transwell assays, and LDH release. Pyroptosis-related proteins were detected by Western blot. MeRIP-qPCR was conducted to measure the m6A level. Finally, the effect of FTO on tumor growth and cisplatin (DDP) sensitivity of OC was checked in a mouse model. We found that FTO was significantly downregulated in OC tissues compared to adjacent normal tissues. In addition, the protein level of FTO was much lower in cisplatin (DDP)-resistant cells than in DDP-sensitive cells. Then, FTO overexpression in DDP-resistant cells decreased IC50 of DDP, increased cell apoptosis, and inhibited cell proliferation, migration, and invasion, whereas FTO silencing cells showed the opposite phenotypes. Moreover, FTO promoted the protein expressions of pyroptosis-related proteins, including NLRP3, cleaved-caspase-1, and GSDMD-NT, to enhance DDP sensitivity, as indicated by lower IC50. Furthermore, FTO activated NLRP3/Caspase-1/GSDMD-mediated pyroptosis in DDP-resistant cells. Mechanically, FTO successfully reduced the total m6A level and NLRP3 mRNA m6A modification to further increase DDP sensitivity in resistant cells. Finally, FTO overexpression promoted DDP-sensitivity and NLRP3 expression to inhibit tumorigenesis in a nude mice xenograft model. Together, our study inferred that FTO triggered NLRP3/Caspase-1/GSDMD-dependent pyroptosis by mediating NLRP3 m6A demethylation to enhance DDP-sensitivity and inhibit the progression of OC. This project indicates that FTO overexpression may be a potential strategy to overcome DDP resistance in OC.
Collapse
Affiliation(s)
- Fubin Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Tianhong Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Lei Gan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Yongming Du
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Lixiao Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Huiqing Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Xueqin Chen
- Department of Traditional Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China.
| | - Yutao Guan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China.
| |
Collapse
|
2
|
Zhang X, Chen J, Zhou X, Zhou D, Liao L, Zhao Y, Wu P, Nie F, Liao Z, Cai Z, Duan X. Exploring diagnostic m6A regulators in primary open-angle glaucoma: insight from gene signature and possible mechanisms by which key genes function. BMC Med Genomics 2025; 18:57. [PMID: 40128732 PMCID: PMC11931807 DOI: 10.1186/s12920-025-02123-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
PURPOSE The purpose of this study was to interrogate the potential role of N6-methyladenosine (m6A) regulators in the process of trabecular meshwork (TM) tissue damage in patients with primary open-angle glaucoma (POAG). METHODS Firstly, the expression profile of m6A regulators in TM tissues of POAG patients was comprehensively analyzed by bioinformatics analysis; Plasmid transfection and siRNA gene interference were used to enhance or weaken the expression levels of YTHDC2 in human trabecular meshwork cells (HTMCs); Cell migration ability was detected by transwell chamber assay; Immunofluorescence staining assay was used to evaluate the expression of extracellular matrix (ECM) related proteins. RESULTS Through the analysis of GSE27276 database, 5 m6A regulators with different expression in POAG were screened out. The results of random forest model showed that these 5 m6A regulators exhibited diagnostic potential and were characteristic genes of POAG. All POAG samples could be effectively divided into two groups based on the expression levels of these 5 hub m6A regulators. Immune cell infiltration analysis indicated that the levels of activated CD8+ T cells and regulatory T cells were different in the two subtypes. HTMC oxidative stress cell model and TGF-β2 stimulation cell model were further constructed to verify the expression of the aforementioned hub m6A regulators, and it was found that YTHDC2 mRNA showed the same expression trend in both models. The silencing of YTHDC2 enhanced the migration ability of HTMCs and increased the synthesis ability of ECM. However, when YTHDC2ΔYTH, which lacks the YTH domain, is overexpressed in HTMCs, there is no significant change in the ECM synthesis ability. CONCLUSIONS The differentially expressed m6A regulators in TM tissues may serve as potential diagnostic biomarkers for POAG. And, in HTMCs, the expression level of YTHDC2 mRNA was changed under oxidative stress or TGF-β2 intervention, and then exerted its regulation on cell migration and ECM synthesis capability through m6A modification, which may be an important part of the disease process of POAG.
Collapse
Affiliation(s)
- Xinyue Zhang
- Hunan Engineering Research Center for Glaucoma with Artificial Intelligence in Diagnosis and Application of New Materials, Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, China
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Jiawei Chen
- Hunan Engineering Research Center for Glaucoma with Artificial Intelligence in Diagnosis and Application of New Materials, Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, China
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Xiaoyu Zhou
- Hunan Engineering Research Center for Glaucoma with Artificial Intelligence in Diagnosis and Application of New Materials, Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, China
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Dengming Zhou
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Liao
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Yang Zhao
- Hunan Engineering Research Center for Glaucoma with Artificial Intelligence in Diagnosis and Application of New Materials, Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, China
| | - Ping Wu
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Fen Nie
- Hunan Engineering Research Center for Glaucoma with Artificial Intelligence in Diagnosis and Application of New Materials, Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, China
| | - Zhimin Liao
- Hunan Engineering Research Center for Glaucoma with Artificial Intelligence in Diagnosis and Application of New Materials, Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, China
| | - Ziyan Cai
- Department of Ophthalmology, The Second Affiliated Hospital of Suzhou University, Suzhou, Jiangsu, China
| | - Xuanchu Duan
- Hunan Engineering Research Center for Glaucoma with Artificial Intelligence in Diagnosis and Application of New Materials, Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, China.
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Wojtukiewicz MZ, Mysliwiec M, Tokajuk A, Kruszewska J, Politynska B, Jamroze A, Wojtukiewicz AM, Tang DG, Honn KV. Tissue factor pathway inhibitor-2 (TFPI-2)-an underappreciated partaker in cancer and metastasis. Cancer Metastasis Rev 2024; 43:1185-1204. [PMID: 39153052 PMCID: PMC11554837 DOI: 10.1007/s10555-024-10205-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024]
Abstract
The coagulation system is known to play an important role in cancer development and metastasis, but the precise mechanisms by which it does so remain incompletely understood. With this in mind, we provide an updated overview of the effects of TFPI-2, a protease inhibitor, on cancer development and metastasis. TFPI-2 interacts with the thrombin cascade and also employs other mechanisms to suppress cancer growth and dissemination, which include extracellular matrix stabilization, promotion of caspase-mediated cell apoptosis, inhibition of angiogenesis and transduction of intracellular signals. Down-regulation of TFPI-2 expression is well documented in numerous types of neoplasms, mainly via promoter methylation. However, the exact role of TFPI-2 in cancer progression and possible approaches to up-regulate TFPI-2 expression warrant further studies. Strategies to reactivate TFPI-2 may represent a promising direction for future anticancer studies and therapy development.
Collapse
Affiliation(s)
- Marek Z Wojtukiewicz
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland.
- Department of Clinical Oncology, Comprehensive Cancer Center of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland.
| | - Marta Mysliwiec
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland
| | - Anna Tokajuk
- Department of Clinical Oncology, Comprehensive Cancer Center of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland
| | - Joanna Kruszewska
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland
| | - Barbara Politynska
- Department of Psychology and Philosophy, Medical University of Bialystok, 37 Szpitalna, 15-295, Bialystok, Poland
- Robinson College, University of Cambridge, Grange Road, Cambridge, CB3 9AN, UK
| | - Anmbreen Jamroze
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Anna M Wojtukiewicz
- Department of Psychology and Philosophy, Medical University of Bialystok, 37 Szpitalna, 15-295, Bialystok, Poland
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Kenneth V Honn
- Department of Pathology-School of Medicine, Bioactive Lipids Research Program, Wayne State University, 540 East Canfield Avenue, Detroit, MI, 48201, USA
- Karmanos Cancer Institute, 4100 John R St, Detroit, MI, 48201, USA
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit, MI, 48202, USA
| |
Collapse
|
4
|
Cai M, Li X, Luan X, Zhao P, Sun Q. Exploring m6A methylation in skin Cancer: Insights into molecular mechanisms and treatment. Cell Signal 2024; 124:111420. [PMID: 39304098 DOI: 10.1016/j.cellsig.2024.111420] [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: 07/15/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
N6-methyladenosine (m6A) is the most common and prevalent internal mRNA modification in eukaryotes. m6A modification is a dynamic and reversible process regulated by methyltransferases, demethylases, and m6A binding proteins. Skin cancers, including melanoma and nonmelanoma skin cancers (NMSCs), are among the most commonly diagnosed cancers worldwide. m6A methylation is involved in the regulation of RNA splicing, translation, degradation, stability, translocation, export, and folding. Aberrant m6A modification participates in the pathophysiological processes of skin cancers and is associated with tumor cell proliferation, invasion, migration, and metastasis during cancer progression. In this review, we provide a comprehensive summary of the biological functions of m6A and the most up-to-date evidence related to m6A RNA modification in skin cancer. We also emphasize the potential clinical applications in the diagnosis and treatment of skin cancers.
Collapse
Affiliation(s)
- Mingjun Cai
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan 250012, Shandong, China
| | - Xueqing Li
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan 250012, Shandong, China
| | - Xueyu Luan
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan 250012, Shandong, China
| | - Pengyuan Zhao
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan 250012, Shandong, China
| | - Qing Sun
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
5
|
Singh S, Gupta S, Abhishek R, Sachan M. Regulation of m 6A (N 6-Methyladenosine) methylation modifiers in solid cancers. Funct Integr Genomics 2024; 24:193. [PMID: 39438339 DOI: 10.1007/s10142-024-01467-z] [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: 07/11/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024]
Abstract
Solid cancers constitute a tremendous burden on global healthcare, requiring a deeper understanding of the molecular mechanisms underlying cancer development and progression. Epigenetic changes, notably N6-methyladenosine (m6A) RNA methylation, have emerged as important contributors to the biology of solid tumors in recent years. This epigenetic mark dynamically affects gene expression at the post-transcriptional level and modulates a variety of cellular processes, making it a focus of research in the context of solid tumors. m6A modification patterns are dysregulated in a variety of solid cancers, including ovarian, breast, lung, colorectal, pancreatic, and others. This dysregulated m6A landscape has been shown to induce significant changes in the expression of oncogenes, tumor suppressors, and genes involved in cancer stem cells, metastasis, and treatment resistance. In solid tumors, the interaction of m6A "writers" (e.g., METTL3, METTL14, and others), "erasers" (e.g., ALKBH5, FTO), and "readers" (e.g., members of YTHDF proteins and others) delicately changes the m6A methylome. Targeting m6A regulators as a potential therapeutic method to control gene expression and prevent tumor development seems a novel strategy. To enhance treatment results, advances in this area of research have led to the development of targeted treatments aiming at restoring or altering m6A alteration patterns in solid tumors.
Collapse
Affiliation(s)
- Sakshi Singh
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Uttar Pradesh, Prayagraj, 211004, India
| | - Sudha Gupta
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Uttar Pradesh, Prayagraj, 211004, India
| | - Rajul Abhishek
- Deparment of Surgical Oncology, Motilal Nehru Medical College, Uttar Pradesh, Prayagraj, 211002, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Uttar Pradesh, Prayagraj, 211004, India.
| |
Collapse
|
6
|
Li R, Kuang Y, Niu Y, Zhang S, Chen S, Su F, Wang J, Lin S, Liu D, Shen C, Liang L, Zheng SG, Jie L, Xiao Y, Xu H. FTO-mediated RNA m 6A methylation regulates synovial aggression and inflammation in rheumatoid arthritis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167341. [PMID: 39025373 DOI: 10.1016/j.bbadis.2024.167341] [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: 04/14/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
Fibroblast-like synoviocytes (FLS) plays an important role in synovial inflammation and joint damage in rheumatoid arthritis (RA). As the most abundant mRNA modification, N6-methyladenosine (m6A) is involved in the development of various diseases; however, its role in RA remains to be defined. In this study, we reported the elevated expression of the m6A demethylase fat mass and obesity-associated protein (FTO) in FLS and synovium from RA patients. Functionally, FTO knockdown or treatment with FB23-2, an inhibitor of the mRNA m6A demethylase FTO, inhibited the migration, invasion and inflammatory response of RA FLS, however, FTO-overexpressed RA FLS exhibited increased migration, invasion and inflammatory response. We further demonstrated that FTO promoted ADAMTS15 mRNA stability in an m6A-IGF2BP1 dependent manner. Notably, the severity of arthritis was significantly reduced in CIA mice with FB23-2 administration or CIA rats with intra-articular injection of FTO shRNA. Our results illustrate the contribution of FTO-mediated m6A modification to joint damage and inflammation in RA and suggest that FTO might be a potential therapeutic target in RA.
Collapse
Affiliation(s)
- Ruiru Li
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Yu Kuang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Yuanyuan Niu
- Department of General Practice, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Shuoyang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Simin Chen
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Fan Su
- Department of Geriatrics, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Jingnan Wang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Di Liu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Chuyu Shen
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Liuqin Liang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Song Jiang Research Institutes, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Ligang Jie
- Department of Rheumatology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Youjun Xiao
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China.
| | - Hanshi Xu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China.
| |
Collapse
|
7
|
Zhang W, Han S, Yuan Y, Xu M, Ding A, Li M. FTO Knockdown-Mediated Maturation of miR-383-5p Inhibits Malignant Advancement of Pancreatic Cancer by Targeting ITGA3. Biochem Genet 2024; 62:2652-2666. [PMID: 38001392 DOI: 10.1007/s10528-023-10560-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023]
Abstract
m6A demethylase FTO is confirmed to be involved in pancreatic cancer progression. FTO regulates miRNA processing. To investigate the regulatory effect of FTO on miR-383-5p and its role in pancreatic cancer. The expression of miR-383-5p, ITGA3, and FTO was predicted using bioinformatic analysis in tissues and was measured using qPCR in cells. Cell biological functions were investigated using MTT assay, Transwell assay, sphere formation assay, and qPCR. The targeting relationship between miR-383-5p and ITGA3 was evaluated using the dual-luciferase reporter assay. The effect of FTO on miR-383-5p processing was evaluated using RIP and MeRIP assay. FTO expression was upregulated in pancreatic cancer and silencing of FTO promoted the processing of miR-383-5p in an m6A-dependent manner. m6A-modified miRNA processing was recognized by IGF2BP1. Downregulation of miR-383-5p reversed FTO knockdown-induced inhibition of cellular processes. The FTO/miR-383-5p/ITGA3 axis facilitated cell viability, metastasis, and stemness in pancreatic cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Radiology, Children's Hospital of Nanjing Medical University, No. 72, Guangzhou Road, Gulou District, Nanjing, 210008, China
| | - Shilong Han
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301, Yanchang Road, Jing'an District, Shanghai, 200072, China
| | - Yifeng Yuan
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301, Yanchang Road, Jing'an District, Shanghai, 200072, China
| | - Minjie Xu
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301, Yanchang Road, Jing'an District, Shanghai, 200072, China
| | - Anle Ding
- AnHui University of Science and Technology, No. 168, Taifeng Road, Huainan, 232001, Anhui, China
| | - Maoquan Li
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, School of Clinical Medicine of Nanjing Medical University, No. 301, Yanchang Road, Jing'an District, Shanghai, 200072, China.
| |
Collapse
|
8
|
Wang K, Mei Z, Zheng M, Liu X, Li D, Wang H. FTO-mediated autophagy inhibition promotes non-small cell lung cancer progression by reducing the stability of SESN2 mRNA. Heliyon 2024; 10:e27571. [PMID: 38495179 PMCID: PMC10943454 DOI: 10.1016/j.heliyon.2024.e27571] [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: 10/22/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024] Open
Abstract
The role of fat mass and obesity-associated protein (FTO), an N6-methyladenosine (m6A) demethylase, in non-small cell lung cancer (NSCLC) has recently received widespread attention. However the underlying mechanisms of FTO-mediated autophagy regulation in NSCLC progression remain elusive. In this study, we found that FTO was significantly upregulated in NSCLC, and downregulation of FTO suppressed the growth, invasion and migration of NSCLC cells by inducing autophagy. FTO knockdown resulted in elevated m6A levels in NSCLC cells. Methylated RNA immunoprecipitation sequencing showed that sestrin 2 (SESN2) was involved in m6A regulation during autophagy in NSCLC cells. Interestingly, m6A modifications in exon 9 of SESN2 regulated its stability. FTO deficiency promoted the binding of insulin-like growth factor 2 mRNA-binding protein 1 to SESN2 mRNA, enhancing its stability and elevating its protein expression. FTO inhibited autophagic flux by downregulating SESN2, thereby promoting the growth, invasion and migration of NSCLC cells. Besides, the mechanism by which FTO blocked SESN2-mediated autophagy activation was associated with the AMPK-mTOR signaling pathway. Taken together, these findings uncover an essential role of the FTO-autophagy-SESN2 axis in NSCLC progression.
Collapse
Affiliation(s)
- Kai Wang
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Zhiqiang Mei
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Meiling Zheng
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Xiaoyan Liu
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Dabing Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Haiyong Wang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| |
Collapse
|
9
|
Zhang FY, Wu L, Zhang TN, Chen HH. KCTD15 acts as an anti-tumor factor in colorectal cancer cells downstream of the demethylase FTO and the m6A reader YTHDF2. Commun Biol 2024; 7:262. [PMID: 38438714 PMCID: PMC10912199 DOI: 10.1038/s42003-024-05880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 02/01/2024] [Indexed: 03/06/2024] Open
Abstract
Potassium Channel Tetramerization Domain Containing 15 (KCTD15) participates in the carcinogenesis of several solid malignancies; however, its role in colorectal cancer (CRC) remains unclear. Here we find that KCTD15 exhibits lower expression in CRC tissues as compared to para-carcinoma tissues. Tetracycline (tet)-induced overexpression and knockdown of KCTD15 confirms KCTD15 as an anti-proliferative and pro-apoptotic factor in CRC both in vitro and in xenografted tumors. N6-methyladenosine (m6A) is known to affect the expression, stabilization, and degradation of RNAs with this modification. We demonstrate that upregulation of fat mass and obesity-associated protein (FTO), a classical m6A eraser, prevents KCTD15 mRNA degradation in CRC cells. Less KCTD15 RNA is recognized by m6A 'reader' YTH N6-Methyladenosine RNA Binding Protein F2 (YTHDF2) in FTO-overexpressed cells. Moreover, KCTD15 overexpression decreases protein expression of histone deacetylase 1 (HDAC1) but increases acetylation of critical tumor suppressor p53 at Lys373 and Lys382. Degradation of p53 is delayed in CRC cells post-KCTD15 overexpression. We further show that the regulatory effects of KCTD15 on p53 are HDAC1-dependent. Collectively, we conclude that KCTD15 functions as an anti-growth factor in CRC cells, and its expression is orchestrated by the FTO-YTHDF2 axis. Enhanced p53 protein stabilization may contribute to KCTD15's actions in CRC cells.
Collapse
Affiliation(s)
- Fang-Yuan Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lin Wu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Huan-Huan Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
10
|
Zhai LL, Li WB, Chen LJ, Wang W, Ju TF, Yin DL. Curcumin inhibits the invasion and migration of pancreatic cancer cells by upregulating TFPI-2 to regulate ERK- and JNK-mediated epithelial-mesenchymal transition. Eur J Nutr 2024; 63:639-651. [PMID: 38129361 DOI: 10.1007/s00394-023-03296-5] [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: 04/29/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE Pancreatic cancer (PC) is one of the most deadly human malignancies. Curcumin is a natural polyphenolic compound with wide-ranging pharmacological effects. Growing evidence suggests that curcumin has anticancer activity against PC, but the mechanism remains incompletely elucidated. This study aimed to investigate the effects and mechanisms of curcumin on the invasion and migration of PC cells. METHODS Effect of curcumin on tissue factor pathway inhibitor (TFPI)-2 mRNA expression in PC cells was initially identified using qRT-PCR. Cytotoxicity of curcumin was assessed with MTT assays and IC50 was calculated. Involvement of ERK and JNK pathways, as well as protein expression of TFPI-2 and epithelial-mesenchymal transition (EMT)-related markers, were detected using immunoblotting. Invasion and migration of PC cells were examined using Transwell assays. TFPI-2 expression was manipulated by transfection with siRNA and shRNA. Rescue assays were used to validate the effect of curcumin on cell invasion and migration via TFPI-2. RESULTS Curcumin increased the expression of TFPI-2 mRNA and protein in PC cells and attenuated cell invasion and migration. Curcumin also inhibited ERK and JNK pathways and EMT in PC cells. Knockdown of TFPI-2 partially reversed the inhibition of ERK and JNK pathways and EMT by curcumin. Mechanistically, curcumin upregulated TFPI-2, thereby inhibiting the ERK and JNK pathways, leading to the inhibition of EMT in PC cells. CONCLUSION Collectively, curcumin inhibits ERK- and JNK-mediated EMT through upregulating TFPI-2, which in turn suppresses the migration and invasion of PC cells. These findings provide new insights into the antitumor mechanism of curcumin.
Collapse
Affiliation(s)
- Lu-Lu Zhai
- Department of General Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, 17 Lujiang Road, Hefei, 230001, People's Republic of China
- Department of General Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 Huansha Road, Hangzhou, 310006, People's Republic of China
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Zhangzhidong Road, Wuhan, 430060, People's Republic of China
| | - Wei-Bo Li
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Zhangzhidong Road, Wuhan, 430060, People's Republic of China
| | - Long-Jiang Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Zhangzhidong Road, Wuhan, 430060, People's Republic of China
| | - Wei Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Zhangzhidong Road, Wuhan, 430060, People's Republic of China
| | - Tong-Fa Ju
- Department of General Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 Huansha Road, Hangzhou, 310006, People's Republic of China.
| | - Da-Long Yin
- Department of General Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, 17 Lujiang Road, Hefei, 230001, People's Republic of China.
| |
Collapse
|
11
|
Wu H, Chen S, Hu Z, Ge R, Ma L, You C, Huang Y. Exploring the prognostic potential of m6A methylation regulators in low-grade glioma: implications for tumor microenvironment modulation. Eur J Med Res 2024; 29:19. [PMID: 38173044 PMCID: PMC10763210 DOI: 10.1186/s40001-023-01621-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/25/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The biological behavior of low-grade glioma (LGG) is significantly affected by N6-methyladenosine (m6A) methylation, an essential epigenetic alteration. Therefore, it is crucial to create a prognostic model for LGG by utilizing genes that regulate m6A methylation. METHODS Using TCGA and GTEx databases. We examined m6A modulator levels in LGG and normal tissues, and investigated PD-L1 and PD-1 expression, immune scores, immune cell infiltration, tumor immune microenvironment (TIME) and potential underlying mechanisms in different LGG clusters. We also performed immunohistochemistry and RT-qPCR to identify essential m6A adjustment factor. RESULTS The results showed that m6A regulatory element expression was significantly increased in LGG tissues and was significantly associated with TMIE. A substantial increase in PD-L1 and PD-1 levels in LGG tissues and high-risk cohorts was observed. PD-L1 expression was positively correlated with FTO, ZCCHC4, and HNRNPD, whereas PD-1 expression was negatively correlated with FTO, ZC3H7B, and HNRNPD. The prognostic signature created using regulators of m6A RNA methylation was shown to be strongly associated with the overall survival of LGG patients, and FTO and ZCCHC4 were confirmed as independent prognostic markers by clinical samples. Furthermore, the results revealed different TIME characteristics between the two groups of patients, indicating disrupted signaling pathways associated with LGG. CONCLUSION Our results present that the m6A regulators play vital role in regulating PD-L1/PD-1 expression and the infiltration of immune cells, thereby exerting a sizable impact on the TIME of LGG. Therefore, m6A regulators have precise predictive value in the prognosis of LGG.
Collapse
Affiliation(s)
- Honggang Wu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Cerebrovascular Disease, The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Siqi Chen
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Ningbo, 315010, Zhejiang, China
| | - Ziliang Hu
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Ningbo, 315010, Zhejiang, China
| | - Rong Ge
- Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315021, China
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yi Huang
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
12
|
Wei H, Xu Y, Lin L, Li Y, Zhu X. A review on the role of RNA methylation in aging-related diseases. Int J Biol Macromol 2024; 254:127769. [PMID: 38287578 DOI: 10.1016/j.ijbiomac.2023.127769] [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: 09/18/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 01/31/2024]
Abstract
Senescence is the underlying mechanism of organism aging and is robustly regulated at the post-transcriptional level. This regulation involves the chemical modifications, of which the RNA methylation is the most common. Recently, a rapidly growing number of studies have demonstrated that methylation is relevant to aging and aging-associated diseases. Owing to the rapid development of detection methods, the understanding on RNA methylation has gone deeper. In this review, we summarize the current understanding on the influence of RNA modification on cellular senescence, with a focus on mRNA methylation in aging-related diseases, and discuss the emerging potential of RNA modification in diagnosis and therapy.
Collapse
Affiliation(s)
- Hong Wei
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China; Department of Neurology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China; Central Laboratory of the Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Yuhao Xu
- Medical School, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Li Lin
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China; Central Laboratory of the Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Yuefeng Li
- Medical School, Jiangsu University, Zhenjiang, Jiangsu 212001, China.
| | - Xiaolan Zhu
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China; Central Laboratory of the Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China.
| |
Collapse
|
13
|
Chen H, Luo W, Lu X, Zhang T. Regulatory role of RNA modifications in the treatment of pancreatic ductal adenocarcinoma (PDAC). Heliyon 2023; 9:e20969. [PMID: 37928039 PMCID: PMC10623179 DOI: 10.1016/j.heliyon.2023.e20969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/22/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely life-threatening malignancy with a relatively unfavorable prognosis. The early occurrence of metastasis and local recurrence subsequent to surgery contribute to the poor survival rates of PDAC patients, thereby limiting the effectiveness of surgical intervention. Additionally, the desmoplastic and immune-suppressive tumor microenvironment of PDAC diminishes its responsiveness to conventional treatment modalities such as chemotherapy, radiotherapy, and immunotherapy. Therefore, it is imperative to identify novel therapeutic targets for PDAC treatment. Chemical modifications are prevalent in various types of RNA and exert significant influence on their structure and functions. RNA modifications, exemplified by m6A, m5C, m1A, and Ψ, have been identified as general regulators of cellular functions. The abundance of specific modifications, such as m6A, has been correlated with cell proliferation, invasion, migration, and patient prognosis in PDAC. Pre-clinical data has indicated that manipulating RNA modification regulators could enhance the efficacy of chemotherapy, radiotherapy, and immunotherapy. Therefore, targeting RNA modifications in conjunction with current adjuvant or neoadjuvant therapy holds promise. The objective of this review is to provide a comprehensive overview of RNA modifications in PDAC treatment, encompassing their behaviors, mechanisms, and potential treatment targets. Therefore, it aims to stimulate the development of novel therapeutic approaches and future clinical trials.
Collapse
Affiliation(s)
- Hao Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyue Lu
- Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Ren X, Tang X, Huang T, Hu Z, Wang Y, Zhou Y. FTO plays a crucial role in gastrointestinal cancer and may be a target for immunotherapy: an updated review. Front Oncol 2023; 13:1241357. [PMID: 37916161 PMCID: PMC10616962 DOI: 10.3389/fonc.2023.1241357] [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: 06/16/2023] [Accepted: 09/22/2023] [Indexed: 11/03/2023] Open
Abstract
Gastrointestinal cancer is a common malignancy with high mortality and poor prognosis. Therefore, developing novel effective markers and therapeutic targets for gastrointestinal cancer is currently a challenging and popular topic in oncology research. Accumulating studies have reported that N6-methyladenosine is the most abundant epigenetic modification in eukaryotes. N6-methyladenosine plays an essential role in regulating RNA expression and metabolism, including splicing, translation, stability, decay, and transport. FTO, the earliest demethylase discovered to maintain the balance of N6-adenosine methylation, is abnormally expressed in many tumors. In this review, we discuss the molecular structure and substrate selectivity of FTO. we focus on the role of FTO in gastrointestinal tumor proliferation, migration, invasion, apoptosis, autophagy, immune microenvironment, and its molecular mechanisms. We also discuss its potential in the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Xiangqing Ren
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaolong Tang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tian Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zenan Hu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
15
|
Zhou J, Tan Q, Tong J, Tong Z, Wang C, Sun B, Fang M, Lv J. PIAS1 upregulation confers protection against Cerulein-induced acute pancreatitis via FTO downregulation by enhancing sumoylation of Foxa2. Genomics 2023; 115:110693. [PMID: 37532089 DOI: 10.1016/j.ygeno.2023.110693] [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: 02/15/2023] [Revised: 07/18/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVE This research discussed the specific mechanism by which PIAS1 affects acute pancreatitis (AP). METHODS PIAS1, Foxa2, and FTO expression was assessed in Cerulein-induced AR42J cells and mice. Loss- and gain-of-function assays and Cerulein induction were conducted in AR42J cells and mice for analysis. The relationship among PIAS1, Foxa2, and FTO was tested. Cell experiments run in triplicate, and eight mice for each animal group. RESULTS Cerulein-induced AP cells and mice had low PIAS1 and Foxa2 and high FTO. Cerulein induced pancreatic injury in mice and inflammation and oxidative stress in pancreatic tissues, which could be reversed by PIAS1 or Foxa2 upregulation or FTO downregulation. PIAS1 elevated SUMO modification of Foxa2 to repress FTO transcription. FTO upregulation neutralized the ameliorative effects of PIAS1 or Foxa2 upregulation on Cerulein-induced AR42J cell injury, inflammation, and oxidative stress. CONCLUSION PIAS1 upregulation diminished FTO transcription by increasing Foxa2 SUMO modification, thereby ameliorating Cerulein-induced AP.
Collapse
Affiliation(s)
- Jiandang Zhou
- Second Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, PR China
| | - Qiao Tan
- Second Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, PR China
| | - Jinxue Tong
- Second Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, PR China
| | - Zhekuan Tong
- Material Supply Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, PR China
| | - Chunlu Wang
- Department of Medical Administration, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, PR China
| | - Bei Sun
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, PR China
| | - Min Fang
- Second Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, PR China
| | - Jiachen Lv
- Second Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, PR China.
| |
Collapse
|
16
|
Tsuji Y, Hara T, Meng S, Sato H, Arao Y, Ofusa K, Ishii H. Role of RNA methylation in the regulation of pancreatic cancer stem cells (Review). Oncol Lett 2023; 26:336. [PMID: 37427348 PMCID: PMC10326658 DOI: 10.3892/ol.2023.13922] [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: 02/02/2023] [Accepted: 04/21/2023] [Indexed: 07/11/2023] Open
Abstract
Pancreatic cancer stem cells (CSCs) play a key role in the initiation and progression of pancreatic adenocarcinoma (PDAC). CSCs are responsible for resistance to chemotherapy and radiation, and for cancer metastasis. Recent studies have indicated that RNA methylation, a type of RNA modification, predominantly occurring as m6A methylation, plays an important role in controlling the stemness of cancer cells, therapeutic resistance against chemotherapy and radiation therapy, and their overall relevance to a patient's prognosis. CSCs regulate various behaviors of cancer through cell-cell communication by secreting factors, through their receptors, and through signal transduction. Recent studies have shown that RNA methylation is involved in the biology of the heterogeneity of PDAC. The present review provides an update on the current understanding of RNA modification-based therapeutic targets against deleterious PDAC. Several key pathways and agents that can specifically target CSCs have been identified, thus providing novel insights into the early diagnosis and efficient treatment of PDAC.
Collapse
Affiliation(s)
- Yoshiko Tsuji
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tomoaki Hara
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Sikun Meng
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hiromichi Sato
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Department of Gastrointestinal Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yasuko Arao
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Ken Ofusa
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Prophoenix Division, Food and Life-Science Laboratory, IDEA Consultants, Inc., Osaka, Osaka 559-8519, Japan
| | - Hideshi Ishii
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
17
|
Ma P, Hao Y, Wang W, Zhang YF, Yu KH, Wang WX. AURKB activates EMT through PI3K/AKT signaling axis to promote ICC progression. Discov Oncol 2023; 14:102. [PMID: 37318676 DOI: 10.1007/s12672-023-00707-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/31/2023] [Indexed: 06/16/2023] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a fatal disease and the molecular mechanism of its progression remains unknown. Aurora Kinase B (AURKB) is a central regulator of chromosome separation and cytokinesis and is abnormally expressed in a variety of cancer cells. This research aimed to explore the effect of AURKB in occurrence and metastasis of ICC. We found that AURKB showed a progressive up-regulation pattern from normal bile duct tissue to ICC with high invasion. Our data showed that AURKB significantly promoted ICC cell proliferation, induced epithelial-mesenchymal transition (EMT), migration and invasion through gain- and loss- of function experiments. In vivo results consistently showed that AURKB up-regulation not only promoted tumor growth, but also promoted tumor metastasis. Importantly, we discovered that AURKB regulates the expressions of EMT-related genes via PI3K/AKT signaling axis. Herein, our results suggest that AURKB induced EMT through the activation of PI3K/AKT signaling pathway is critical to the progression of ICC, which may be a prospective therapeutic treatment for overcoming ICC metastasis and progression.
Collapse
Affiliation(s)
- Peng Ma
- Deportment of Hepatobiliary Surgery, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Ying Hao
- Deportment of Hepatobiliary Surgery, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Wei Wang
- Deportment of Hepatobiliary Surgery, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Yue-Feng Zhang
- Deportment of Hepatobiliary Surgery, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Kai-Huan Yu
- Deportment of Hepatobiliary Surgery, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| | - Wei-Xing Wang
- Deportment of Hepatobiliary Surgery, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
18
|
The Emerging Role of m6A Modification in Endocrine Cancer. Cancers (Basel) 2023; 15:cancers15041033. [PMID: 36831377 PMCID: PMC9954123 DOI: 10.3390/cancers15041033] [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: 12/22/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
With the development of RNA modification research, N6-methyladenosine (m6A) is regarded as one of the most important internal epigenetic modifications of eukaryotic mRNA. It is also regulated by methylase, demethylase, and protein preferentially recognizing the m6A modification. This dynamic and reversible post-transcriptional RNA alteration has steadily become the focus of cancer research. It can increase tumor stem cell self-renewal and cell proliferation. The m6A-modified genes may be the primary focus for cancer breakthroughs. Although some endocrine cancers are rare, they may have a high mortality rate. As a result, it is critical to recognize the significance of endocrine cancers and identify new therapeutic targets that will aid in improving disease treatment and prognosis. We summarized the latest experimental progress in the m6A modification in endocrine cancers and proposed the m6A alteration as a potential diagnostic marker for endocrine malignancies.
Collapse
|