1
|
Wang D, Li H, Wang Q, Liu Y. NAT10 mediated polycystic ovary syndrome through the ac4C modification of CXCL14. J Steroid Biochem Mol Biol 2025; 251:106767. [PMID: 40300667 DOI: 10.1016/j.jsbmb.2025.106767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder among women of reproductive age, underscoring the critical importance of investigating its regulatory mechanisms. N-Acetyltransferase 10 (NAT10) is a crucial enzyme involved in mRNA acetylation modification, mediating target genes expression through N4-acetylcytidine (ac4C) modification to regulate the biological function of various diseases. Nonetheless, the specific role of NAT10 in PCOS regulation remains undisclosed. Ac4C dot hybridization experiment was conducted to determine ac4C expression in PCOS tissues. RT-qPCR was employed to assess the expression levels of NAT10 and CXCL14 in PCOS tissues and KGN cells. Cells viability was assessed using the CCK-8 method, while cell proliferation capacity was evaluated through the colony formation assay and EDU assay. Flow cytometry analysis was utilized to measure the apoptosis rate. The ac4C modification level was determined by acrp-qPCR analysis. RIP and luciferase reporter experiments confirmed the target binding relationship. The rat experiments confirmed the specific regulatory role of NAT10 in polycystic ovary syndrome in vivo. This study highlighted reduced levels of NAT10 and ac4C in PCOS, where silencing NAT10 boosts KGN cell proliferation and suppresses apoptosis. Additionally, NAT10-mediated ac4C modification governed the chemokine CXCL14 expression. Our research unveiled that NAT10 modulated PCOS occurrence and progression by enhancing the CXCL14 mRNA stability through acetylation, presenting potential novel insights into the mechanisms of mRNA acetylation in PCOS.
Collapse
Affiliation(s)
- Ding Wang
- Department of Gynecology, Yichang Maternity & Child Healthcare Hospital, Yichang, Hubei 443001, China.
| | - Hui Li
- Department of Gynecology, Yichang Maternity & Child Healthcare Hospital, Yichang, Hubei 443001, China
| | - Qiaoling Wang
- Department of Gynecology, Yichang Maternity & Child Healthcare Hospital, Yichang, Hubei 443001, China
| | - Yunxia Liu
- Department of Gynecology, Yichang Maternity & Child Healthcare Hospital, Yichang, Hubei 443001, China
| |
Collapse
|
2
|
Zhou X, Liu W, Liang Z, Liang J, Zhang T, Gao W, Yang Z. Key epigenetic enzymes modulated by natural compounds contributes to tumorigenicity. Int J Biol Macromol 2025; 301:140391. [PMID: 39880237 DOI: 10.1016/j.ijbiomac.2025.140391] [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: 12/04/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Dysregulation of epigenetic regulation is observed in numerous tumor cells. The therapeutic effects of natural products on tumors were investigated through a comprehensive analysis of active ingredients derived from various structured natural products. The analysis focuses on regulating key enzymes involved in epigenetic control. To study the modulation of these enzymes for tumor treatment, the structural characteristics of natural products that impact tumorigenesis were identified. The presence of specific patterns suggests that compounds sharing structural similarities can potentially induce therapeutic effects on identical tumors through modulation of distinct modifying enzymes. Structurally analogous natural products can likewise achieve therapeutic effects across diverse tumor types via their interaction with a common epigenetic enzyme. There exist numerous flavonoids with the capability to modulate METTL3, thereby influencing the development of various tumors. The normalization process was implemented to account for a common phenomenon, wherein structurally distinct compounds effectively target the same tumor by modulating a shared key enzyme. By summarizing, valuable insights into the role of compound-epigenetic enzymes in tumor development have been obtained. This discovery establishes a crucial scientific foundation for the prevention and treatment of tumor development through the utilization of structurally similar natural active ingredients.
Collapse
Affiliation(s)
- Xiaoyue Zhou
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wanqing Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ziqi Liang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiali Liang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenyi Gao
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Zizhao Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of General Surgery, Shanghai Seventh People's Hospital, Shanghai 200137, China.
| |
Collapse
|
3
|
Chen Y, Wang J, Xu J, Kou R, Lan B, Qin Z. NAT10 promotes gastric cancer progression by enhancing the N4-acetylcytidine modification of TNC mRNA. Infect Agent Cancer 2025; 20:20. [PMID: 40158090 PMCID: PMC11955120 DOI: 10.1186/s13027-025-00650-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a very aggressive malignant tumor of the digestive system. Previous studies have shown that N-acetyltransferase 10 (NAT10) can regulate the N4-acetylcytidine (ac4C) modification of downstream mRNAs through certain pathways to promote the progression of various tumors. However, reports on the regulatory effects of NAT10 on GC are rare. This study aimed to explore the potential mechanism by which NAT10 regulated GC progression. METHODS Clinical samples were used to study the correlation between NAT10 expression and poor prognosis in patients with GC by univariate analysis and multivariate analysis. In vitro and in vivo assays were performed to assess the effects of NAT10 and Tenascin C (TNC) on the malignant biological behaviors of GC cells. Acetylated RNA immunoprecipitation sequencing was conducted to explore the role of NAT10 in ac4C modification in GC. mRNA stability and translation efficiency assays were performed to investigate the effect of changes in NAT10 expression on its target TNC. RESULTS Analysis of clinical samples revealed that NAT10 expression was abnormally elevated and positively correlated with TNC expression in GC, and increased NAT10 expression led to poor overall survival. In vitro and in vivo experiments revealed that high NAT10 expression promoted the invasive and proliferative capacity of GC cells. Rescue experiments suggested that TNC played an important role in the above process. Mechanistically, the acetylation-based RNA immunoprecipitation sequencing and acetylated RNA immunoprecipitation qPCR results indicated that NAT10 regulated the level of ac4C modification by binding to specific regions in TNC mRNA, increasing mRNA stability and translation, upregulating TNC expression, further activating the TNC/Akt/TGF-β1 positive feedback loop. CONCLUSIONS In summary, our results reveal that NAT10 plays a critical role in GC development by affecting TNC mRNA stability and translation efficiency, which ultimately activates the TNC/Akt/TGF-β1 positive feedback loop. This study is expected to provide a novel target and theoretical basis for improving the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Yu Chen
- Department of Gastrointestinal Surgery 2 Section, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Gastrointestinal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jinzhou Wang
- Department of Gastrointestinal Surgery 2 Section, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Gastrointestinal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jiuhua Xu
- Department of Gastrointestinal Surgery 2 Section, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Gastrointestinal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Ruilong Kou
- Department of Gastrointestinal Surgery 2 Section, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Gastrointestinal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Bin Lan
- Department of Gastrointestinal Surgery 2 Section, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Gastrointestinal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| | - Zhiwei Qin
- Department of Gastrointestinal Surgery 2 Section, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Gastrointestinal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
4
|
Yao L, Xie P, Dong D, Guo Y, Guan J, Zhang W, Chung CR, Zhao Z, Chiang YC, Lee TY. Caps-ac4C: An effective computational framework for identifying N4-acetylcytidine sites in human mRNA based on deep learning. J Mol Biol 2025; 437:168961. [PMID: 39884569 DOI: 10.1016/j.jmb.2025.168961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
N4-acetylcytidine (ac4C) is a crucial post-transcriptional modification in human mRNA, involving the acetylation of the nitrogen atom at the fourth position of cytidine. This modification, catalyzed by N-acetyltransferases such as NAT10, is primarily found in mRNA's coding regions and enhances translation efficiency and mRNA stability. ac4C is closely associated with various diseases, including cancer. Therefore, accurately identifying ac4C in human mRNA is essential for gaining deeper insights into disease pathogenesis and provides potential pathways for the development of novel medical interventions. In silico methods for identifying ac4C are gaining increasing attention due to their cost-effectiveness, requiring minimal human and material resources. In this study, we propose an efficient and accurate computational framework, Caps-ac4C, for the precise detection of ac4C in human mRNA. Caps-ac4C utilizes chaos game representation to encode RNA sequences into "images" and employs capsule networks to learn global and local features from these RNA "images". Experimental results demonstrate that Caps-ac4C achieves state-of-the-art performance, achieving 95.47% accuracy and 0.912 MCC on the test set, surpassing the current best methods by 10.69% accuracy and 0.216 MCC. In summary, Caps-ac4C represents the most accurate tool for predicting ac4C sites in human mRNA, highlighting its significant contribution to RNA modification research. For user convenience, we developed a user-friendly web server, which can be accessed for free at:https://awi.cuhk.edu.cn/~Caps-ac4C/index.php.
Collapse
Affiliation(s)
- Lantian Yao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China; School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China.
| | - Peilin Xie
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China; School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China
| | - Danhong Dong
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China
| | - Yilin Guo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China
| | - Jiahui Guan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China; School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China
| | - Wenyang Zhang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China
| | - Chia-Ru Chung
- Department of Computer Science and Information Engineering, National Central University, Taoyuan, Taiwan
| | - Zhihao Zhao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China
| | - Ying-Chih Chiang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China; School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, 518172 Shenzhen, China.
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
| |
Collapse
|
5
|
Xiao N, Liu H, Zhang R, Li Y, Zhao X, Chen H, Zhang C, Zhu Y, Lu H, Wang X, Liu H, Wan J. N-acetyltransferase 10 impedes EZH2/H3K27me3/GABARAP axis mediated autophagy and facilitates lung cancer tumorigenesis through enhancing SGK2 mRNA acetylation. Int J Biol Macromol 2025; 297:139823. [PMID: 39814292 DOI: 10.1016/j.ijbiomac.2025.139823] [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: 11/23/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
N4-acetylcytidine (ac4C) is a critical RNA modification implicated in cancer progression. Currently, N-acetyltransferase 10 (NAT10) is recognized as the sole "writer" protein responsible for ac4C modification. However, the study of NAT10 and ac4C modification in lung cancer remains sparse. In this study, we observed a significant upregulation of NAT10 expression in lung cancer, which is strongly correlated with poor prognostic outcomes. In vitro and in vivo experiments have demonstrated that NAT10 facilitates the proliferation, migration, and invasion of non-small cell lung cancer (NSCLC) cells while inhibiting autophagy flux. Mechanistically, NAT10 may enhance mRNA stability through ac4c modification at the 3' untranslated region (UTR) of SGK2 mRNA. Furthermore, SGK2 interacts with EZH2 and phosphorylates it at threonine 367, leading to increased protein stability of EZH2 and a reduction in its ubiquitination. Additionally, NAT10 impedes autophagy flux by preventing the fusion of autophagosomes with lysosomes and suppressing GABARAP transcription, which is regulated by EZH2-mediated H3K27me3. In summary, our study elucidates the biological significance and molecular mechanisms of the NAT10/SGK2/EZH2 axis in the pathogenesis of lung cancer, potentially providing novel prognostic markers and therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Nan Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruike Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangzhuan Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huanxiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chenxing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongshen Lu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuanzhi Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongchun Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
6
|
Wang K, Hu Y, Nie J, Zeng Q, Hu Y, Wu H. Chicken hnRNPK suppresses interferon production, thereby enhancing IBDV replication. Res Vet Sci 2025; 184:105527. [PMID: 39765197 DOI: 10.1016/j.rvsc.2025.105527] [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: 11/03/2024] [Revised: 12/29/2024] [Accepted: 01/01/2025] [Indexed: 02/01/2025]
Abstract
Heterogeneous ribonucleoprotein K (hnRNPK) is a well-known RNA-binding protein initially identified for its role in inhibiting the growth of various human tumors. Members of the hnRNP family have also been implicated in both interferon production and RNA virus replication. However, the role of chicken hnRNPK (chhnRNPK) in the replication of Infectious Bursal Disease Virus (IBDV) remains unclear. In this study, we identified chhnRNPK as a protein that interacts with genomic double-stranded RNA (dsRNA). Following IBDV infection, chhnRNPK was recruited to the virus replication complex in the cytoplasm. Furthermore, chhnRNPK expression inhibited dsRNA-induced interferon production, specifically at the mitochondrial antiviral signaling protein (MAVS) step. Overexpression of chhnRNPK significantly enhanced virus replication, while knockdown of chhnRNPK increased dsRNA-induced interferon production and subsequently disrupted IBDV replication. Collectively, these findings suggest that chhnRNPK promotes IBDV replication by interacting with genomic dsRNA, highlighting a novel host factor that regulates viral replication.
Collapse
Affiliation(s)
- Ke Wang
- Department of Veterinary Preventive Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Zhimin Street, Qingshan Lake, Nanchang 330045, PR China
| | - Ying Hu
- Department of Veterinary Preventive Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Zhimin Street, Qingshan Lake, Nanchang 330045, PR China
| | - Jiangjiang Nie
- Department of Veterinary Preventive Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Zhimin Street, Qingshan Lake, Nanchang 330045, PR China
| | - Qinghua Zeng
- Department of Veterinary Preventive Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Zhimin Street, Qingshan Lake, Nanchang 330045, PR China
| | - Yu Hu
- Department of Veterinary Preventive Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Zhimin Street, Qingshan Lake, Nanchang 330045, PR China
| | - Huansheng Wu
- Department of Veterinary Preventive Medicine, College of Animal Science and Technology, Jiangxi Agricultural University, Zhimin Street, Qingshan Lake, Nanchang 330045, PR China.
| |
Collapse
|
7
|
Liang P, Zhou D, Liao J, Lai W, Sheng X, Wang Y. N-acetyltransferase 10 Promotes Cervical Cancer Progression Via N4-acetylation of SLC7A5 mRNA. FRONT BIOSCI-LANDMRK 2025; 30:26756. [PMID: 40018938 DOI: 10.31083/fbl26756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/18/2024] [Accepted: 12/25/2024] [Indexed: 03/01/2025]
Abstract
INTRODUCTION N-acetyltransferase 10 (NAT10) mediates N4-acetylcytidine (ac4C) mRNA modification and promotes malignant tumor progression. However, there has been limited research on its role in cervical cancer. This study aimed to decipher the role of NAT10 in cervical cancer. METHODS The prognostic value of NAT10 was explored using the cancer genome atlas (TCGA) database and immunohistochemistry of cervical cancer tissue. The biological actions of NAT10 in cervical cancer were investigated by cell proliferation, transwell, wound healing, and chicken chorioallantoic membrane assays. The therapeutic action of remodelin (a NAT10 inhibitor) was verified in a nude mouse model. Mechanistic analyses were conducted by RNA sequencing, ac4C dot blotting, acetylated RNA immunoprecipitation, quantitative PCR, and RNA stability experiments. RESULTS NAT10 was overexpressed in cervical carcinoma and its overexpression was associated with poor prognosis. NAT10 knockout impaired proliferative and metastatic potentials of cervical cancer cells, while its overexpression had the opposite effects. Remodelin impaired cervical cancer proliferation in vivo and in vitro. NAT10 acetylated solute carrier family 7 member 5 (SLC7A5) enhanced mRNA stability to regulate SLC7A5 expression. CONCLUSIONS NAT10 exerts a critical role in cervical cancer progression via acetylating SLC7A5 mRNA and could represent a key prognostic and therapeutic target in cervical cancer.
Collapse
Affiliation(s)
- Peili Liang
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, 510260 Guangzhou, Guangdong, China
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital, Guangzhou Medical University, 510150 Guangzhou, Guangdong, China
| | - Dongmei Zhou
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital, Guangzhou Medical University, 510150 Guangzhou, Guangdong, China
| | - Jinrong Liao
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, 510260 Guangzhou, Guangdong, China
| | - Wujiang Lai
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, 510260 Guangzhou, Guangdong, China
- Department of Obstetrics and Gynecology, Shunde Hospital, The First People's Hospital of Shunde, Southern Medical University, 528300 Foshan, Guangdong, China
| | - Xiujie Sheng
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital, Guangzhou Medical University, 510150 Guangzhou, Guangdong, China
| | - Yifeng Wang
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, 510260 Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Chen Y, Yang J, Du Y, Yan Z, Gao J, Zhang H, Wu Q, Nian B, Huang X, Da M. Acetyltransferase NAT10 promotes gastric cancer progression by regulating the Wnt/β-catenin signaling pathway and enhances chemotherapy resistance. Discov Oncol 2025; 16:173. [PMID: 39945932 PMCID: PMC11825422 DOI: 10.1007/s12672-025-01917-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND N-acetyltransferase 10 (NAT10) is involved in several cellular processes. NAT10 expression is essential for the promotion of mRNA translation and stability. In some situations, deregulation of NAT10 has been attributed to the development of multiple types of cancer. NAT10 is significantly upregulated in various gastrointestinal tumors, including esophageal, colorectal, pancreatic, and liver cancers, and is correlated with poor prognosis. Additionally, NAT10 expression contributes to chemotherapy resistance in both esophageal and colorectal cancers. Nevertheless, the role of NAT10 in gastric cancer (GC), a type of gastrointestinal tumor, is not fully understood. METHODS Throughout this investigation, our team evaluated NAT10 expression levels in GC patient samples and databases available to the general public. Based on the knockdown and overexpression of NAT10, in vitro experiments were conducted to examine the effects of NAT10 on GC progression and resistance to chemotherapy. RESULTS Our study demonstrated that GC tissues exhibit increased levels of NAT10. Downregulation of NAT10 decreased GC cell proliferation, migration, and invasiveness. Conversely, upregulation of NAT10 resulted in the opposite effect. Furthermore, NAT10 fosters the progression of GC cells by activating the Wnt/β-catenin signaling pathway. NAT10 also promotes resistance to cisplatin chemotherapy. CONCLUSIONS Our findings indicated that expression of NAT10 promoted GC progression through activation of the Wnt/β-catenin signaling pathway. We investigated the effect of NAT10 on the viability of GC cells treated with different doses of cisplatin. The results showed that NAT10 expression could impact the effectiveness of chemotherapy resistance in GC. This implies that using NAT10 as a target may be a potential therapeutic strategy for treating GC.
Collapse
Affiliation(s)
- Yawen Chen
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Jian Yang
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Yadan Du
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Zaihua Yan
- The Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Jieyun Gao
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, 730000, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Haoyang Zhang
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Qiong Wu
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Bowen Nian
- Department of General Surgery, The First People's Hospital of Huating, Huating, 744100, China
| | - Xiujuan Huang
- Department of Hematology, Gansu Provincial Hospital, Lanzhou, 730000, China
- Department of Hematology, Shanxi Provincial People's Hospital, Xian, 710068, China
| | - Mingxu Da
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China.
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
9
|
Song Y, Cheng M. NAT10 promotes ovarian cancer cell migration, invasion, and stemness via N4-acetylcytidine modification of CAPRIN1. BMC Womens Health 2025; 25:54. [PMID: 39923057 PMCID: PMC11806784 DOI: 10.1186/s12905-025-03567-9] [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: 08/20/2024] [Accepted: 01/16/2025] [Indexed: 02/10/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological tumor. N4-acetylcytidine (ac4C) modification, catalyzed by the acetyltransferase NAT10, is involved in the occurrence and development of cancers. This study aimed to investigate the role of NAT10 in OC and the underlying molecular mechanisms. The expression of NAT10 and CAPRIN1 in OC cells lines were measured using quantitative real-time polymerase chain reaction and immunoblotting. Biological behaviors of OC cells were evaluated using EdU, Transwell, sphere formation, and immunoblotting assays. The molecular mechanism of NAT10 function was analyzed using bioinformatics, ac4C- RNA immunoprecipitation, and actinomycin D treatment assay. The effect of NAT10 on OC progression in vivo was evaluated using xenograft tumor model. The results indicated that NAT10 and CAPRIN1 were highly expressed in OC cells. NAT10 knockdown suppressed OC cell proliferation, migration, invasiveness, stemness, and epithelial-mesenchymal transition in vitro, and impeded tumor growth in vivo. Additionally, CAPRIN1 expression was found to be positively related to NAT10 expression in OC. Silencing of NAT10 inhibited ac4C levels of CAPRIN1 and reduced its RNA stability. Moreover, overexpression of CAPRIN1 reversed the suppression of migration, invasion, and stemness caused by NAT10 knockdown, while knockdown of CAPRIN1 alone inhibited these malignant behaviors of OC cells. In conclusion, NAT10 promotes OC progression by promoting cellular migration, invasion, and stemness via upregulating CAPRIN1 expression. Mechanistically, NAT10 stabilizes CAPRIN1 by promoting its ac4C modification. These findings suggest that NAT10 may be a promising therapy target for OC.
Collapse
Affiliation(s)
- Yang Song
- Department of Gynecologic Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, P.R. China
| | - Min Cheng
- Department of Gynecologic Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, P.R. China.
| |
Collapse
|
10
|
Xiao B, Wu S, Tian Y, Huang W, Chen G, Luo D, Cai Y, Chen M, Zhang Y, Liu C, Zhao J, Li L. Advances of NAT10 in diseases: insights from dual properties as protein and RNA acetyltransferase. Cell Biol Toxicol 2024; 41:17. [PMID: 39725720 PMCID: PMC11671434 DOI: 10.1007/s10565-024-09962-6] [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: 07/14/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
N-acetyltransferase 10 (NAT10) is a member of the Gcn5-related N-acetyltransferase (GNAT) family and it plays a crucial role in various cellular processes, such as regulation of cell mitosis, post-DNA damage response, autophagy and apoptosis regulation, ribosome biogenesis, RNA modification, and other related pathways through its intrinsic protein acetyltransferase and RNA acetyltransferase activities. Moreover, NAT10 is closely associated with the pathogenesis of tumors, Hutchinson-Gilford progeria syndrome (HGPS), systemic lupus erythematosus, pulmonary fibrosis, depression and host-pathogen interactions. In recent years, mRNA acetylation has emerged as a prominent focus of research due to its pivotal role in regulating RNA stability and translation. NAT10 stands out as the sole identified modification enzyme responsible for RNA acetylation. There remains some ambiguity regarding the similarities and differences in NAT10's actions on protein and RNA substrates. While NAT10 involves acetylation modification in both cases, which is a crucial molecular mechanism in epigenetic regulation, there are significant disparities in the catalytic mechanisms, regulatory pathways, and biological processes involved. Therefore, this review aims to offer a comprehensive overview of NAT10 as a protein and RNA acetyltransferase, covering its basic catalytic features, biological functions, and roles in related diseases.
Collapse
Affiliation(s)
- Bin Xiao
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China.
- Department of Laboratory Medicine, Guangdong Provincial Second Hospital of Traditional Chinese Medicine, Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine, The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510095, Guangdong, China.
| | - Shunhong Wu
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Yan Tian
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Weikai Huang
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Guangzhan Chen
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Dongxin Luo
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Yishen Cai
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Ming Chen
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Yuqian Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Chuyan Liu
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Junxiu Zhao
- College of Public Health, Dali University, Dali, 671003, Yunnan, China
| | - Linhai Li
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China.
| |
Collapse
|
11
|
Chen Y, Xue G, Hu Y, Da M. Exploring the role of NAT10 in regulating EMT and JAK2/STAT3 pathway activation in cervical cancer progression. Asian J Surg 2024:S1015-9584(24)02836-7. [PMID: 39676018 DOI: 10.1016/j.asjsur.2024.11.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/11/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024] Open
Affiliation(s)
- Yawen Chen
- Gansu University of Chinese Medicine, Lanzhou, 730000, China; Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Gaimei Xue
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yonghua Hu
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Mingxu Da
- Gansu University of Chinese Medicine, Lanzhou, 730000, China; Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China.
| |
Collapse
|
12
|
Tang B, Wang K, Ren Q, Zhou J, Xu Y, Liu L, Yin B, Zhang Y, Huang Q, Lv R, Luo Z, Zhao H, Shen L. GALNT14-mediated O-glycosylation drives lung adenocarcinoma progression by reducing endogenous reactive oxygen species generation. Cell Signal 2024; 124:111477. [PMID: 39426495 DOI: 10.1016/j.cellsig.2024.111477] [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: 07/18/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
Aberrant glycosylation, resulting from dysregulated expression of glycosyltransferases, is a prevalent feature of cancer cells. N-acetylgalactosaminyltransferase-14 (GALNT14) serves as a pivotal enzyme responsible for initiating O-GalNAcylation. It remains unclear whether and how GALNT14 affects lung adenocarcinoma (LUAD). Here, GALNT14 expression in LUAD was analyzed by searching public databases and verified by examining clinical samples. Bioinformatics, LC-MS/MS, RNA-seq, and RIP-seq analyses were used to uncover the mechanism underlying GALNT14. We observed that GALNT14 was frequently overexpressed in LUAD tissues. High GALNT14 expression was positively associated with advanced TNM stage, larger tumor size, and unfavorable prognosis. Functionally, GALNT14 facilitated LUAD cell proliferation, migration, and invasion in vitro and accelerated tumor growth in vivo. Mechanistically, GALNT14 reduced the accumulation of endogenous reactive oxygen species (ROS) to exert its oncogenic function via O-glycosylating hnRNPUL1 to upregulate AKR1C2 expression. Meanwhile, GALNT14 expression was directly modulated by miR-125a.These findings indicated that GALNT14-mediated O-GalNAcylation could drive LUAD progression via eliminating ROS and might be a valuable therapeutic target.
Collapse
Affiliation(s)
- Bingbing Tang
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China; Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Kelong Wang
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China; Key Laboratory of Cancer Therapy Resistance and Clinical Translational Study, Shiyan 442000, Hubei, China
| | - Qiulei Ren
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Junshuo Zhou
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yuewen Xu
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Liaoyuan Liu
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Bin Yin
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yaling Zhang
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Qian Huang
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Ruiqi Lv
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Zhiguo Luo
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China; Key Laboratory of Cancer Therapy Resistance and Clinical Translational Study, Shiyan 442000, Hubei, China.
| | - Hongyan Zhao
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China.
| | - Li Shen
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China; Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China; Key Laboratory of Cancer Therapy Resistance and Clinical Translational Study, Shiyan 442000, Hubei, China.
| |
Collapse
|
13
|
Gu Z, Zou L, Pan X, Yu Y, Liu Y, Zhang Z, Liu J, Mao S, Zhang J, Guo C, Li W, Geng J, Zhang W, Yao X, Shen B. The role and mechanism of NAT10-mediated ac4C modification in tumor development and progression. MedComm (Beijing) 2024; 5:e70026. [PMID: 39640362 PMCID: PMC11617596 DOI: 10.1002/mco2.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
RNA modification has emerged as a crucial area of research in epigenetics, significantly influencing tumor biology by regulating RNA metabolism. N-acetyltransferase 10 (NAT10)-mediated N4-acetylcytidine (ac4C) modification, the sole known acetylation in eukaryotic RNA, influences cancer pathogenesis and progression. NAT10 is the only writer of ac4C and catalyzes acetyl transfer on targeted RNA, and ac4C helps to improve the stability and translational efficiency of ac4C-modified RNA. NAT10 is highly expressed and associated with poor prognosis in pan-cancers. Based on its molecular mechanism and biological functions, ac4C is a central factor in tumorigenesis, tumor progression, drug resistance, and tumor immune escape. Despite the increasing focus on ac4C, the specific regulatory mechanisms of ac4C in cancer remain elusive. The present review thoroughly analyzes the current knowledge on NAT10-mediated ac4C modification in cancer, highlighting its broad regulatory influence on targeted gene expression and tumor biology. This review also summarizes the limitations and perspectives of current research on NAT10 and ac4C in cancer, to identify new therapeutic targets and advance cancer treatment strategies.
Collapse
Affiliation(s)
- Zhuoran Gu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Libin Zou
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Xinjian Pan
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Yang Yu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Yongqiang Liu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Zhijin Zhang
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Ji Liu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Shiyu Mao
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Junfeng Zhang
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Changcheng Guo
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Wei Li
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Jiang Geng
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Wentao Zhang
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Xudong Yao
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Bing Shen
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of MedicineTongi UniversityShanahaiChina
| |
Collapse
|
14
|
Ouyang W, Huang Z, Wan K, Nie T, Chen H, Yao H. RNA ac 4C modification in cancer: Unraveling multifaceted roles and promising therapeutic horizons. Cancer Lett 2024; 601:217159. [PMID: 39128536 DOI: 10.1016/j.canlet.2024.217159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/18/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
RNA modifications play a crucial role in cancer development, profoundly influencing various stages of the RNA lifecycle. These stages encompass nuclear processing, nuclear export, splicing, and translation in the cytoplasm. Among RNA modifications, RNA ac4C modification, also known as N4-acetylcytidine, stands out for its unique role in acetylation processes. Specific proteins regulate RNA ac4C modification, maintaining the dynamic and reversible nature of these changes. This review explores the molecular mechanisms and biological functions of RNA ac4C modification. It examines the intricate ways in which RNA ac4C modification influences the pathogenesis and progression of cancer. Additionally, the review provides an integrated overview of the current methodologies for detecting RNA ac4C modification. Exploring the potential applications of manipulating this modification suggests avenues for novel therapeutic strategies, potentially leading to more effective cancer treatments in the future.
Collapse
Affiliation(s)
- Wenhao Ouyang
- Department of Oncology, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China
| | - Zhenjun Huang
- Department of Oncology, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China
| | - Keyu Wan
- The First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Tiantian Nie
- The First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Haizhu Chen
- Department of Oncology, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China.
| | - Herui Yao
- Department of Oncology, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China.
| |
Collapse
|
15
|
Beiki H, Sturgill D, Arango D, Relier S, Schiffers S, Oberdoerffer S. Detection of ac4C in human mRNA is preserved upon data reassessment. Mol Cell 2024; 84:1611-1625.e3. [PMID: 38640896 PMCID: PMC11353019 DOI: 10.1016/j.molcel.2024.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/19/2023] [Accepted: 03/18/2024] [Indexed: 04/21/2024]
Abstract
We recently reported the distribution of N4-acetylcytidine (ac4C) in HeLa mRNA at base resolution through chemical reduction and the induction of C:T mismatches in sequencing (RedaC:T-seq). Our results contradicted an earlier report from Schwartz and colleagues utilizing a similar method termed ac4C-seq. Here, we revisit both datasets and reaffirm our findings. Through RedaC:T-seq reanalysis, we establish a low basal error rate at unmodified nucleotides that is not skewed to any specific mismatch type and a prominent increase in C:T substitutions as the dominant mismatch type in both treated wild-type replicates, with a high degree of reproducibility across replicates. In contrast, through ac4C-seq reanalysis, we uncover significant data quality issues including insufficient depth, with one wild-type replicate yielding 2.7 million reads, inconsistencies in reduction efficiencies between replicates, and an overall increase in mismatches involving thymine that could obscure ac4C detection. These analyses bolster the detection of ac4C in HeLa mRNA through RedaC:T-seq.
Collapse
Affiliation(s)
- Hamid Beiki
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - David Sturgill
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Daniel Arango
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sebastien Relier
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sarah Schiffers
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Shalini Oberdoerffer
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Dalhat MH, Narayan S, Serio H, Arango D. Dissecting the oncogenic properties of essential RNA-modifying enzymes: a focus on NAT10. Oncogene 2024; 43:1077-1086. [PMID: 38409550 PMCID: PMC11092965 DOI: 10.1038/s41388-024-02975-9] [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: 12/26/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/28/2024]
Abstract
Chemical modifications of ribonucleotides significantly alter the physicochemical properties and functions of RNA. Initially perceived as static and essential marks in ribosomal RNA (rRNA) and transfer RNA (tRNA), recent discoveries unveiled a dynamic landscape of RNA modifications in messenger RNA (mRNA) and other regulatory RNAs. These findings spurred extensive efforts to map the distribution and function of RNA modifications, aiming to elucidate their distribution and functional significance in normal cellular homeostasis and pathological states. Significant dysregulation of RNA modifications is extensively documented in cancers, accentuating the potential of RNA-modifying enzymes as therapeutic targets. However, the essential role of several RNA-modifying enzymes in normal physiological functions raises concerns about potential side effects. A notable example is N-acetyltransferase 10 (NAT10), which is responsible for acetylating cytidines in RNA. While emerging evidence positions NAT10 as an oncogenic factor and a potential target in various cancer types, its essential role in normal cellular processes complicates the development of targeted therapies. This review aims to comprehensively analyze the essential and oncogenic properties of NAT10. We discuss its crucial role in normal cell biology and aging alongside its contribution to cancer development and progression. We advocate for agnostic approaches to disentangling the intertwined essential and oncogenic functions of RNA-modifying enzymes. Such approaches are crucial for understanding the full spectrum of RNA-modifying enzymes and imperative for designing effective and safe therapeutic strategies.
Collapse
Affiliation(s)
- Mahmood H Dalhat
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Sharath Narayan
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, USA
| | - Hannah Serio
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Daniel Arango
- Department of Pharmacology, Northwestern University, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
17
|
Amin R, Ha NH, Qiu T, Holewinski R, Lam KC, Lopès A, Liu H, Tran AD, Lee MP, Gamage ST, Andresson T, Goldszmid RS, Meier JL, Hunter KW. Loss of NAT10 disrupts enhancer organization via p300 mislocalization and suppresses transcription of genes necessary for metastasis progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.577116. [PMID: 38410432 PMCID: PMC10896336 DOI: 10.1101/2024.01.24.577116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Acetylation of protein and RNA represent a critical event for development and cancer progression. NAT10 is the only known RNA acetylase that catalyzes the N4-actylcytidine (ac4C) modification of RNAs. Here, we show that the loss of NAT10 significantly decreases lung metastasis in allograft and genetically engineered mouse models of breast cancer. NAT10 interacts with a mechanosensitive, metastasis susceptibility protein complex at the nuclear pore. In addition to its canonical role in RNA acetylation, we find that NAT10 interacts with p300 at gene enhancers. NAT10 loss is associated with p300 mislocalization into heterochromatin regions. NAT10 depletion disrupts enhancer organization, leading to alteration of gene transcription necessary for metastatic progression, including reduced myeloid cell-recruiting chemokines that results in a less metastasis-prone tumor microenvironment. Our study uncovers a distinct role of NAT10 in enhancer organization of metastatic tumor cells and suggests its involvement in the tumor-immune crosstalk dictating metastatic outcomes.
Collapse
|
18
|
Zhang S, Liu Y, Ma X, Gao X, Ru Y, Hu X, Gu X. Recent advances in the potential role of RNA N4-acetylcytidine in cancer progression. Cell Commun Signal 2024; 22:49. [PMID: 38233930 PMCID: PMC10795262 DOI: 10.1186/s12964-023-01417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/02/2023] [Indexed: 01/19/2024] Open
Abstract
N4-acetylcytidine (ac4C) is a highly conserved chemical modification widely found in eukaryotic and prokaryotic RNA, such as tRNA, rRNA, and mRNA. This modification is significantly associated with various human diseases, especially cancer, and its formation depends on the catalytic activity of N-acetyltransferase 10 (NAT10), the only known protein that produces ac4C. This review discusses the detection techniques and regulatory mechanisms of ac4C and summarizes ac4C correlation with tumor occurrence, development, prognosis, and drug therapy. It also comments on a new biomarker for early tumor diagnosis and prognosis prediction and a new target for tumor therapy. Video Abstract.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yafeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Xiao Ma
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaohui Gao
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yi Ru
- Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Xinjun Hu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, 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.
| |
Collapse
|
19
|
Wang L, Tao Y, Zhai J, Xue M, Zheng C, Hu H. The emerging roles of ac4C acetylation "writer" NAT10 in tumorigenesis: A comprehensive review. Int J Biol Macromol 2024; 254:127789. [PMID: 37926318 DOI: 10.1016/j.ijbiomac.2023.127789] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
The quick progress of epigenetic study has kindled new hope for treating many cancers. When it comes to RNA epigenetics, the ac4C acetylation modification is showing promise, whereas N-acetyltransferase 10 plays a wide range of biological functions, has a significant impact on cellular life events, and is frequently highly expressed in many malignant tumors. N-acetyltransferase 10 is an acetyltransferase with important biological involvement in cellular processes and lifespan. Because it is highly expressed in many malignant tumors, it is considered a pro-carcinogenic gene. The review aims to introduce NAT10, summarize the effects of ac4C acetylation on tumor growth from multiple angles, and discuss the possible therapeutic targeting of NAT10 and the future directions of ac4C acetylation investigations.
Collapse
Affiliation(s)
- Leisheng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China; Wuxi Medical College, Jiangnan University, Wuxi, 214122, China
| | - Yue Tao
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China; Wuxi Medical College, Jiangnan University, Wuxi, 214122, China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, 028000, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, Henan, China, 450001
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Hao Hu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China; Wuxi Medical College, Jiangnan University, Wuxi, 214122, China; Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China; Hepatobiliary and Pancreatic Surgery, The Third Hospital Affiliated to Nantong University, Wuxi, 214041, China; Medical School, Nantong University, Nantong, 226001, China; Wuxi Institute of Hepatobiliary Surgery, Wuxi, 214122, China
| |
Collapse
|
20
|
Rodrigues P, Bangali H, Ali E, Nauryzbaevish AS, Hjazi A, Fenjan MN, Alawadi A, Alsaalamy A, Alasheqi MQ, Mustafa YF. The mechanistic role of NAT10 in cancer: Unraveling the enigmatic web of oncogenic signaling. Pathol Res Pract 2024; 253:154990. [PMID: 38056132 DOI: 10.1016/j.prp.2023.154990] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
N-acetyltransferase 10 (NAT10), a versatile enzyme, has gained considerable attention as a significant player in the complex realm of cancer biology. Its enigmatic role in tumorigenesis extends across a wide array of cellular processes, impacting cell growth, differentiation, survival, and genomic stability. Within the intricate network of oncogenic signaling, NAT10 emerges as a crucial agent in multiple cancer types, such as breast, lung, colorectal, and leukemia. This compelling research addresses the intricate complexity of the mechanistic role of NAT10 in cancer development. By elucidating its active participation in essential physiological processes, we investigate the regulatory role of NAT10 in cell cycle checkpoints, coordination of chromatin remodeling, and detailed modulation of the delicate balance between apoptosis and cell survival. Perturbations in NAT10 expression and function have been linked to oncogenesis, metastasis, and drug resistance in a variety of cancer types. Furthermore, the bewildering interactions between NAT10 and key oncogenic factors, such as p53 and c-Myc, are deciphered, providing profound insights into the molecular underpinnings of cancer pathogenesis. Equally intriguing, the paradoxical role of NAT10 as a potential tumor suppressor or oncogene is influenced by context-dependent factors and the cellular microenvironment. This study explores the fascinating interplay of genetic changes, epigenetic changes, and post-translational modifications that shape the dual character of NAT10, revealing the delicate balance between cancer initiation and suppression. Taken together, this overview delves deeply into the enigmatic role of NAT10 in cancer, elucidating its multifaceted roles and its complex interplay with oncogenic networks.
Collapse
Affiliation(s)
- Paul Rodrigues
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Saudi Arabia.
| | - Harun Bangali
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Saudi Arabia
| | - Eyhab Ali
- College of Chemistry, Al-Zahraa University for Women, Karbala, Iraq
| | - Abdreshov Serik Nauryzbaevish
- Institute of Genetics and Physiology SC MSHE RK, Laboratory of Physiology Lymphatic System, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohammed N Fenjan
- College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Ahmed Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Iraq
| | - Ali Alsaalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|