1
|
Choi JC. Perinuclear organelle trauma at the nexus of cardiomyopathy pathogenesis arising from loss of function LMNA mutation. Nucleus 2025; 16:2449500. [PMID: 39789731 PMCID: PMC11730615 DOI: 10.1080/19491034.2024.2449500] [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: 10/31/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Over the past 25 years, nuclear envelope (NE) perturbations have been reported in various experimental models with mutations in the LMNA gene. Although the hypothesis that NE perturbations from LMNA mutations are a fundamental feature of striated muscle damage has garnered wide acceptance, the molecular sequalae provoked by the NE damage and how they underlie disease pathogenesis such as cardiomyopathy (LMNA cardiomyopathy) remain poorly understood. We recently shed light on one such consequence, by employing a cardiomyocyte-specific Lmna deletion in vivo in the adult heart. We observed extensive NE perturbations prior to cardiac function deterioration with collateral damage in the perinuclear space. The Golgi is particularly affected, leading to cytoprotective stress responses that are likely disrupted by the progressive deterioration of the Golgi itself. In this review, we discuss the etiology of LMNA cardiomyopathy with perinuclear 'organelle trauma' as the nexus between NE damage and disease pathogenesis.
Collapse
Affiliation(s)
- Jason C. Choi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Zhang A, Yang J, Wang M, Li Y, Hu T, Xie J, Xu Y, Cao W. Target inhibition of NAT10-mediated ac4C modification prevents seizure behavior in mice. Neuropharmacology 2025; 272:110415. [PMID: 40107603 DOI: 10.1016/j.neuropharm.2025.110415] [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: 12/18/2024] [Revised: 02/24/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
N4-acetylation of Cytidine (ac4C), catalyzed by its only known enzyme N-acetyltransferase 10 (NAT10), facilitates cellular mRNA translation and stability, but its function in brain disorders especially epilepsy is poorly understood. By using pentylenetetrazole (PTZ) induced mouse model of epilepsy, we first displayed spatiotemporally expression of ac4C and NAT10 in the mouse brain. To corroborate the alteration of ac4C and NAT10 in epilepsy, we used acute PTZ, chronic PTZ and intrahippocampal kainic acid (IHKA) mouse model. We then utilized a combination of viral tool and pharmacological approaches to implicate NAT10 mediated ac4C modification in seizure behaviors. We found that the expression of ac4C was increased in epileptic brain tissues in mouse models of epilepsy, which might be due to the up-regulated NAT10. Block of NAT10 led to both reduced brain ac4C level and resistance to PTZ or KA-induced seizure behavior, while hippocampal over-expression of NAT10 causes exacerbated seizure behavior. In support of such a role, our data demonstrated that the loss or gains of ac4C modification could normalize or exacerbate neuronal over-activation in epileptic brain tissues, respectively. Mechanically, we observed that block the NAT10 or over-expression NAT10 lead to reduced or enhanced BDNF, respectively. While the BDNF pathway inhibitor rescued the hippocampal NAT10 over-expression induced aggravated seizure behavior in the chronic PTZ treated mice. Therefore, our work provides the first demonstration of the ac4C levels in an epilepsy mice model, targeted to prevent ac4C by NAT10 inhibition seems to be effective in preventing and treating epilepsy.
Collapse
Affiliation(s)
- Aomei Zhang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jingwen Yang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Meng Wang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yujia Li
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tao Hu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jialing Xie
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yang Xu
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wenyu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
3
|
Znaidi R, Massiani-Beaudoin O, Mailly P, Monnet H, Bonnifet T, Joshi RL, Fuchs J. Nuclear translocation of the LINE-1 encoded ORF1 protein alters nuclear envelope integrity in human neurons. Brain Res 2025; 1857:149579. [PMID: 40157412 DOI: 10.1016/j.brainres.2025.149579] [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: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
LINE-1 retrotransposons are increasingly implicated in aging and neurodegenerative diseases, yet the precise pathogenic mechanisms remain elusive. While the endonuclease and reverse transcriptase activities of LINE-1-encoded ORF2p can induce DNA damage and inflammation, a role of LINE-1 ORF1p in cellular dysfunctions stays unassigned. Here we demonstrate, using a neuronal cellular model, that ORF1p translocates into the nucleus upon arsenite-induced stress, directly interacting with nuclear import (KPNB1), nuclear pore complex (NUP153), and nuclear lamina (Lamin B1) proteins. Nuclear translocation of ORF1p disrupts nuclear integrity, nucleocytoplasmic transport, and heterochromatin structure, features linked to neurodegeneration and aging. Elevated nuclear ORF1p levels induced either by arsenite-induced stress, ORF1p overexpression, or as observed in Parkinson's disease post-mortem brain tissues correlate with impaired nuclear envelope (NE) morphology. Stress-induced nuclear alterations are mitigated by blocking ORF1p nuclear import or with the anti-aging drug remodelin. This study thus reveals a pathogenic action of nuclear ORF1p in human neurons driving NE alterations and thereby contributing to LINE-1-mediated cell toxicity.
Collapse
Affiliation(s)
- Rania Znaidi
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | | | - Philippe Mailly
- Orion Imaging Facility, CIRB, Collège de France, Université PSL, CNRS, INSERM, Labex Memolife, 75005 Paris, France
| | - Héloïse Monnet
- Orion Imaging Facility, CIRB, Collège de France, Université PSL, CNRS, INSERM, Labex Memolife, 75005 Paris, France
| | - Tom Bonnifet
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Rajiv L Joshi
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France.
| | - Julia Fuchs
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France.
| |
Collapse
|
4
|
Song A, Liu B, Li W, Chen B, Gui P, Zhang H, Zhu C, Xu Y, Jiang T, Song J. Competitive binding between DDX21 and SIRT7 enhances NAT10-mediated ac 4C modification to promote colorectal cancer metastasis and angiogenesis- DDX21 promotes colorectal cancer metastasis. Cell Death Dis 2025; 16:353. [PMID: 40301349 PMCID: PMC12041575 DOI: 10.1038/s41419-025-07656-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 05/01/2025]
Abstract
DExD- box helicase 21 (DDX21) is overexpressed in colorectal cancer (CRC) and is positively correlated with poor prognosis and the malignant phenotype of CRC. Functional characterization indicated that DDX21 promotes CRC metastasis and angiogenesis both in vitro and in vivo. N-acetyltransferase 10 (NAT10) is a key regulator of the N4-acetylcytidine (ac4C) modification of mRNA, regulating the stabilization of mRNA via ac4C modification. Here, we identified that DDX21 competitive binding with sirtuin 7 (SIRT7), inducing the overexpression of NAT10. Furthermore, DDX21 upregulates NAT10 expression to enhance ac4C modification and the stability of ATAD2, SOX4 and SNX5 mRNAs, which mediate CRC metastasis and angiogenesis. Overall, the present study revealed a mechanism of DDX21/NAT10-mediated mRNA stability in CRC, laying the foundation for the use of DDX21 as a therapeutic target to overcome metastasis and angiogenesis in CRC. DDX21 competitive binding with sirtuin 7 (SIRT7), inducing the overexpression of NAT10. Furthermore, DDX21 upregulates NAT10 expression to enhance ac4C modification and the stability of ATAD2, SOX4 and SNX5 mRNAs, which mediate CRC metastasis and angiogenesis.
Collapse
Affiliation(s)
- Angxi Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Bowen Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenjing Li
- Central Laboratory, Xuzhou NO.1 people's hospital, Xuzhou, China
| | - Bingyuan Chen
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
| | - Pengkun Gui
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
| | - Hao Zhang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Can Zhu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China.
| | - Jun Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
5
|
Wang JN, Suo XG, Yu JT, Luo QC, Ji ML, Zhang MM, Zhu Q, Cheng XR, Hou C, Chen X, Wang F, Xu CH, Li C, Xie SS, Wei J, Zhang DF, Zhang XR, Wang ZJ, Dong YH, Zhu S, Peng LJ, Li XY, Chen HY, Xu T, Jin J, Chen FX, Meng XM. NAT10 exacerbates acute renal inflammation by enhancing N4-acetylcytidine modification of the CCL2/CXCL1 axis. Proc Natl Acad Sci U S A 2025; 122:e2418409122. [PMID: 40261924 PMCID: PMC12054813 DOI: 10.1073/pnas.2418409122] [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: 09/12/2024] [Accepted: 03/08/2025] [Indexed: 04/24/2025] Open
Abstract
Inflammation plays an essential role in eliminating microbial pathogens and repairing tissues, while sustained inflammation accelerates kidney damage and disease progression. Therefore, understanding the mechanisms of the inflammatory response is vital for developing therapies for inflammatory kidney diseases like acute kidney injury (AKI), which currently lacks effective treatment. Here, we identified N-acetyltransferase 10 (NAT10) as an important regulator for acute inflammation. NAT10, the only known "writer" protein for N4-acetylcytidine (ac4C) acetylation, is elevated in renal tubules across various AKI models, human biopsies, and cultured tubular epithelial cells (TECs). Conditional knockout (cKO) of NAT10 in mouse kidneys attenuates renal dysfunction, inflammation, and infiltration of macrophages and neutrophils, whereas its conditional knock-in (cKI) exacerbates these effects. Mechanistically, our findings from ac4C-RIP-seq and RNA-seq analyses revealed that NAT10-mediated ac4C acetylation enhances the mRNA stability of a range of key chemokines, including C-C motif chemokine ligand 2 (CCL2) and C-X-C motif chemokine ligand 1(CXCL1), promoting macrophage and neutrophil recruitment and accelerating renal inflammation. Additionally, CCL2 and CXCL1 neutralizing antibodies or their receptor inhibitors, abrogated renal inflammation in NAT10-overexpression TECs or NAT10-cKI mice. Importantly, inhibiting NAT10, either through Adeno-associated virus 9 (AAV9)-mediated silencing or pharmacologically with our found inhibitor Cpd-155, significantly reduces renal inflammation and injury. Thus, targeting the NAT10/CCL2/CXCL1 axis presents a promising therapeutic strategy for treating inflammatory kidney diseases.
Collapse
Affiliation(s)
- Jia-nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Xiao-guo Suo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Ju-tao Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Qi-chao Luo
- School of Basic Medicine, Anhui Medical University, Hefei230032, China
| | - Ming-lu Ji
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Meng-meng Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Qi Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Xin-ran Cheng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Chao Hou
- School of Basic Medicine, Anhui Medical University, Hefei230032, China
| | - Xin Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Fang Wang
- Department of Pharmacy, Lu’an Hospital of Anhui Medical University, Lu’an People’s Hospital of Anhui Province, Lu’an237006, China
| | - Chuan-hui Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Chao Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Shuai-shuai Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Jie Wei
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei230601, Anhui, China
| | - Dan-feng Zhang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei230601, Anhui, China
| | - Xin-ru Zhang
- School of Basic Medicine, Anhui Medical University, Hefei230032, China
| | - Zhi-juan Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei230601, Anhui, China
| | - Yu-hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Sai Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Li-jin Peng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Xiang-yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Hai-yong Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong999077, Hong Kong
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| | - Juan Jin
- School of Basic Medicine, Anhui Medical University, Hefei230032, China
| | - Fei Xavier Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Xiao-ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei230032, China
| |
Collapse
|
6
|
Ji HN, Zhou HQ, Qie JB, Lu WM, Gao HT, Wu DH. Dysregulated ac4C modification of mRNA in a mouse model of early-stage Alzheimer's disease. Cell Biosci 2025; 15:45. [PMID: 40223095 PMCID: PMC11995559 DOI: 10.1186/s13578-025-01389-8] [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: 11/26/2024] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND The identification and intervention of Alzheimer's Disease (AD) in its early-stage allows for the timely implementation of lifestyle modifications and therapeutic strategies. Although dysregulation of protein expression has been reported in the brain from AD patients and AD animal models, the underlying mechanisms remain poorly understood. N4-acetylcytidine (ac4C), the only known form of RNA acetylation in eukaryotes, has recently been shown to regulate mRNA stability and translation efficiency. However, the dysregulation of ac4C associated with abnormal protein expression levels in the brain of early-stage mouse models of AD remains to be elucidated. METHODS This study investigated ac4C modifications, mRNA and protein expression in the hippocampus of 3 and 6-month-old 5×FAD mice, a mouse model of AD, and wild-type (WT) littermates. The multi-omics analysis was performed: acetylated RNA immunoprecipitation followed by next-generation sequencing (acRIP-seq) to identify ac4C mRNAs, deep RNA sequencing (RNA-seq) to quantify mRNA abundance, and label-free quantitative proteomics to assess protein expression levels. In addition, we used acRIP-qPCR, regular qPCR and western blots to verify the ac4C, mRNA and protein levels of some key genes that were identified by the high-throughput assays. RESULTS Proteomic analysis revealed significant change of protein expression in the hippocampus of 3-months-old 5×FAD mice, compared with WT littermates. In contrast, RNA-seq analysis indicated that there were no substantial alterations in mRNA expression levels in the hippocampus of 3-months-old 5×FAD mice, compared to WT littermates. Strikingly, acRIP-seq revealed notable variations in ac4C modification on mRNAs, particularly those associated with synaptic structure and function, in the hippocampus of 3-months-old 5×FAD mice, compared with WT littermates. The ac4C modifications were found to be correlated with protein expression changes. Genes that are essential for synaptic function and cognition, including GRIN1, MAP2, and DNAJC6, exhibited reduced ac4C and protein levels in 3-months-old 5×FAD mice, without any corresponding changes in the mRNA levels, compared with WT littermates. Moreover, only a small part of dysregulated ac4C mRNAs identified in the 3-month-old 5×FAD mice were found in the 6-month-old 5×FAD mice. CONCLUSIONS Altogether these results identified abnormal ac4C modification of mRNAs that may contribute to the dysregulation of protein synthesis in the hippocampus from an early-stage mouse model of AD.
Collapse
Affiliation(s)
- Hao-Nan Ji
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Hai-Qian Zhou
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Jing-Bo Qie
- Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Wen-Mei Lu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Hai-Tao Gao
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Dan-Hong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China.
- Center of Community-Based Health Research, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Chen C, Wang Z, Lin Q, Li M, Xu L, Fu Y, Zhao X, Ma Z, Xu J, Zhou S, Zhang M, Qian Y, Bao L, Wang B, Wang M, Ding Q, Wang Q, Wang S. NAT10 Promotes Gastric Cancer Liver Metastasis by Modulation of M2 Macrophage Polarization and Metastatic Tumor Cell Hepatic Adhesion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410263. [PMID: 39985269 PMCID: PMC12005778 DOI: 10.1002/advs.202410263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/21/2024] [Indexed: 02/24/2025]
Abstract
The relationship between patterns of RNA modifications and gastric cancer (GC) liver metastasis (GCLM) remains unclear. Here, by single-cell sequencing, clinical sample analysis, and mouse model studies, an abnormal increase in the expression of the RNA acetyltransferase N-acetyltransferase 10 (NAT10) in liver metastatic GC cells is identified. NAT10-mediated N4-acetylcytidine modification of CXCL2 and KLF5 mRNA increases their stability. Then, secreted CXCL2 is found to promote the infiltration and polarization of M2-like macrophages to produce oncostatin M, which transcriptionally activates NAT10 expression via STAT3 signaling. In addition, organoid models confirm that NAT10 promotes the adhesion of GC cells to hepatocytes. Mechanistically, KLF5 transcriptionally activates ITGαV, facilitating GC cell attachment to hepatocytes. Intriguingly, high expression of NAT10/KLF5 axis is associated with poor prognosis of GC patients and targeting this axis significantly reduces GCLM in preclinical murine models. Collectively, these findings suggest the clinical significance of NAT10 in developing targeted therapies for GC patients with liver metastasis.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
| | - Zhangding Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
| | - Qingfeng Lin
- Department of OncologyJiangyin Clinical College of Xuzhou Medical UniversityJiangyin Hospital Affiliated to Nantong UniversityJiangyin People's HospitalJiangyin214400China
| | - Mengmeng Li
- Medical School of Nanjing UniversityNanjing210093China
| | - Lei Xu
- Department of GastroenterologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Yao Fu
- Department of PathologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Xiaoya Zhao
- Medical School of Nanjing UniversityNanjing210093China
| | - Zhuang Ma
- Medical School of Nanjing UniversityNanjing210093China
| | - Jiawen Xu
- Medical School of Nanjing UniversityNanjing210093China
| | - Shimeng Zhou
- Medical School of Nanjing UniversityNanjing210093China
| | - Mingyue Zhang
- Medical School of Nanjing UniversityNanjing210093China
| | - Yun Qian
- Medical School of Nanjing UniversityNanjing210093China
| | - Linsen Bao
- Division of Gastric SurgeryDepartment of General SurgeryThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Bo Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
| | - Meng Wang
- Division of Gastric SurgeryDepartment of General SurgeryThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Qingqing Ding
- Department of Geriatric OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
| | - Shouyu Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
- Medical School of Nanjing UniversityNanjing210093China
| |
Collapse
|
8
|
Chen JF, Xu P, Cai WL, Chen H, Wingrove E, Shi X, Li W, Biancon G, Zhang M, Balabaki A, Krop ED, Asare E, Zhang Y, Yin M, Tebaldi T, Meier JL, Westbrook TF, Halene S, Liu Y, Shen H, Nguyen DX, Yan Q. An in vivo screen identifies NAT10 as a master regulator of brain metastasis. SCIENCE ADVANCES 2025; 11:eads6021. [PMID: 40138393 PMCID: PMC11939035 DOI: 10.1126/sciadv.ads6021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025]
Abstract
Emerging evidence has shown that epigenetic regulation plays a fundamental role in cancer metastasis, the major cause of cancer-related deaths. Here, we conducted an in vivo screen for vulnerabilities of brain metastasis and identified N-acetyltransferase 10 (NAT10) as a driver of brain metastasis. Knockdown of NAT10 restrains cancer cell proliferation and migration in vitro and tumor growth and brain metastasis in vivo. The poorly characterized RNA helicase domain of NAT10 is critical for cell growth in vitro, while both RNA helicase and NAT domains are essential for primary tumor growth and brain metastasis in vivo. Mechanically, NAT10 promotes the expression of 3-phosphoglycerate dehydrogenase (PHGDH) and phosphoserine aminotransferase 1 (PSAT1), two enzymes for serine biosynthesis implicated in brain metastasis. Silencing PHGDH or PSAT1 in metastatic breast cancer cells inhibits their growth in the serine/glycine-limited condition, phenocopying the effects of NAT10 depletion. These findings establish NAT10 as a key regulator of brain metastasis and nominate NAT10 as a target for treating metastasis.
Collapse
Affiliation(s)
- Jocelyn F. Chen
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Peng Xu
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Wesley L. Cai
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Huacui Chen
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Emily Wingrove
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Xiaojian Shi
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
- Systems Biology Institute, Yale West Campus, West Haven, CT 06516, USA
| | - Wenxue Li
- Department of Pharmacology, Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Giulia Biancon
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Internal Medicine (Section of Hematology), Yale School of Medicine, New Haven, CT 06520, USA
| | - Meiling Zhang
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Amer Balabaki
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Ethan D. Krop
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Elianna Asare
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yangyi Zhang
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mingzhu Yin
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Toma Tebaldi
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Internal Medicine (Section of Hematology), Yale School of Medicine, New Haven, CT 06520, USA
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Jordan L. Meier
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Thomas F. Westbrook
- Therapeutic Innovation Center (THINC), Verna & Marrs McLean Department of Biochemistry & Molecular Pharmacology, Department of Molecular and Human Genetics, and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephanie Halene
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Internal Medicine (Section of Hematology), Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yansheng Liu
- Department of Pharmacology, Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Biomedical Informatics & Data Science, Yale School of Medicine, New Haven, CT 06510, USA
| | - Hongying Shen
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
- Systems Biology Institute, Yale West Campus, West Haven, CT 06516, USA
| | - Don X. Nguyen
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Internal Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, CT 06520, USA
| | - Qin Yan
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
9
|
Debnath TK, Abell NS, Li YR, Devanathan SK, Navedo E, Xhemalçe B. NAT10 and N4-acetylcytidine restrain R-loop levels and related inflammatory responses. SCIENCE ADVANCES 2025; 11:eads6144. [PMID: 40138394 PMCID: PMC11939041 DOI: 10.1126/sciadv.ads6144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
N4-acetylcytidine (ac4C) is deposited on diverse RNAs by N-acetyltransferase 10 (NAT10), a protein with high biological relevance for aging and cancer. We performed a comprehensive survey of ac4C using metabolic labeling, sodium cyanoborohydride chemical treatment coupled to next-generation sequencing (NGS), and ac4C antibody-based cell and molecular biology techniques. Our analysis shows that NAT10-dependent ac4C-acetylation is robust in rRNA and specific tRNAs but low/spurious in mRNA. It also revealed an inflammatory signature and mutagenesis at transcriptionally active sites in NAT10-KO cells. This finding led us to explore the role of NAT10 in R-loops, which were recently linked to APOBEC3B-mediated mutagenesis. Our analysis showed that R-loops are ac4C-acetylated in a NAT10-dependent manner. Furthermore, NAT10 restrains the levels of R-loops at a subset of differentially expressed genes in a catalytic activity-dependent manner. Together with cellular biology data showing ac4C-modified RNA in endosomal structures, we propose that increased levels of ac4C-unmodified RNAs, likely derived from R-loops, in endosomal structures induce inflammatory responses.
Collapse
Affiliation(s)
- Turja K. Debnath
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
| | - Nathan S. Abell
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
| | - Yi-Ru Li
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
- Winship Cancer Center and Department of Biochemistry, Emory University School of Medicine, Wayne O Rollins Research Center, 1510 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Sravan K. Devanathan
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
- Winship Cancer Center and Department of Biochemistry, Emory University School of Medicine, Wayne O Rollins Research Center, 1510 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Enrique Navedo
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
- Winship Cancer Center and Department of Biochemistry, Emory University School of Medicine, Wayne O Rollins Research Center, 1510 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Blerta Xhemalçe
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
- Winship Cancer Center and Department of Biochemistry, Emory University School of Medicine, Wayne O Rollins Research Center, 1510 Clifton Rd NE, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Wan X, Wang L, Khan MA, Peng L, Sun X, Yi X, Wang Z, Chen K. NAT10-mediated N4-acetylcytidine modification in KLF9 mRNA promotes adipogenesis. Cell Death Differ 2025:10.1038/s41418-025-01483-x. [PMID: 40123006 DOI: 10.1038/s41418-025-01483-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 02/14/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Dysfunctional adipogenesis is a major contributor of obesity. N-acetyltransferase 10 (NAT10) plays a crucial role in regulating N4-acetylcysteine (ac4C) modification in tRNA, 18SrRNA, and mRNA. As the sole "writer" in the ac4C modification process, NAT10 enhances mRNA stability and translation efficiency. There are few reports on the relationship between NAT10 and adipogenesis, as well as obesity. Our study revealed a significant upregulation of NAT10 in adipose tissues of obese individuals and high-fat diet-fed mice. Furthermore, our findings revealed that the overexpression of NAT10 promotes adipogenesis, while its silencing inhibits adipogenesis in both human adipose tissue-derived stem cells (hADSCs) and 3T3-L1 cells. These results indicate the intimate relationship between NAT10 and obesity. After silencing mouse NAT10 (mNAT10), we identified 30 genes that exhibited both hypo-ac4C modification and downregulation in their expression, utilizing a combined approach of acRIP-sequencing (acRIP-seq) and RNA-sequencing (RNA-seq). Among these genes, we validated KLF9 as a target of NAT10 through acRIP-PCR. KLF9, a pivotal transcription factor that positively regulates adipogenesis. Our findings showed that NAT10 enhances the stability of KLF9 mRNA and further activates the CEBPA/B-PPARG pathway. Furthermore, a dual-luciferase reporter assay demonstrated that NAT10 can bind to three motifs of mouse KLF9 and one motif of human KLF9. In vivo studies revealed that adipose tissue-targeted mouse AAV-NAT10 (AAV-shRNA-mNAT10) inhibits adipose tissue expansion in mice. Additionally, Remodelin, a specific NAT10 inhibitor, significantly reduced body weight, adipocyte size, and adipose tissue expansion in high-fat diet-fed mice by inhibiting KLF9 mRNA ac4C modification. These findings provide novel insights and experimental evidence of the prevention and treatment of obesity, highlighting NAT10 and its downstream targets as potential therapeutic targets.
Collapse
Affiliation(s)
- Xinxing Wan
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Linghao Wang
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Md Asaduzzaman Khan
- Department of Biochemistry and Microbiology, School of Health & Life Sciences, North South University, Dhaka, Bangladesh
| | - Lin Peng
- Department of Nephrology, The First Hospital of Changsha, Changsha, Hunan, PR China
| | - Xiaoying Sun
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Xuan Yi
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Zhouqi Wang
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Ke Chen
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
11
|
Huang L, Lu Y, He R, Guo X, Zhou J, Fu Z, Li J, Liu J, Chen R, Zhou Y, Zhou Q. N 4-acetylcytidine modification of ITGB5 mRNA mediated by NAT10 promotes perineural invasion in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res 2025; 44:103. [PMID: 40119353 PMCID: PMC11929334 DOI: 10.1186/s13046-025-03362-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/08/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Perineural invasion (PNI) is a hallmark feature of pancreatic ductal adenocarcinoma (PDAC), which occurs at a high incidence and significantly contributes to PDAC lethality and poor survival. Despite its prevalence and association with poor prognosis, the molecular mechanisms underlying PNI in PDAC remain unclear. METHODS We investigated clinical samples from two cohorts by UPLC/MS-MS to profiled significantly altered chemical RNA modifications in PDAC tissues with PNI lesions. Dorsal root ganglion coculture systems and sciatic nerve injection models validated PNI ability. We combined RNA-seq, acRIP-seq and ac4C-seq with CRISPR-based techniques to explore the regulatory mechanism of ac4C modification on the integrin beta 5 (ITGB5) transcript. RESULT We reported that N4-acetylcytidine (ac4C) is a significantly altered chemical RNA modification in PDAC tissues with PNI lesions. In vitro and in vivo models demonstrated that tumor cells overexpression of N-acetyltransferase 10 (NAT10), the writer enzyme of mRNA ac4C modification, enhances PNI in PDAC. Further analysis revealed decreased ac4C levels on transcripts of the focal adhesion pathway, particular on ITGB5, in NAT10-knockdown PDAC cells. This ac4C modification in the CDS region of ITGB5 mRNA promotes its stability, subsequently activating the ITGB5-pFAK-pSrc pathway. CRISPR-based analysis further confirmed the crucial role of NAT10-mediated ac4C modification in regulating ITGB5 expression. Combining small-molecule inhibitors targeting NAT10 and focal adhesion kinase (FAK) significantly attenuated PNI in vivo. CONCLUSION Our findings reveal a previously unrecognized ac4C-mediated epigenetic mechanism in PNI and propose a novel therapeutic strategy to improve survival in PDAC patients. NAT10 promotes PNI via ac4C modification in PDAC.
Collapse
Affiliation(s)
- Leyi Huang
- Department of Pancreas Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, People's Republic of China
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Yanan Lu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Rihua He
- Department of Pancreas Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Xiaofeng Guo
- Guangzhou Digestive Disease Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Jiajia Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, People's Republic of China
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Zhiqiang Fu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, People's Republic of China
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Jingwen Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510275, People's Republic of China
| | - Jianping Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, People's Republic of China.
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, People's Republic of China.
| | - Rufu Chen
- Department of Pancreas Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, People's Republic of China.
| | - Yu Zhou
- Department of Pancreas Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, People's Republic of China.
| | - Quanbo Zhou
- Department of Pancreas Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510080, People's Republic of China.
| |
Collapse
|
12
|
Xu Z, Zhu M, Geng L, Zhang J, Xia J, Wang Q, An H, Xia A, Yu Y, Liu S, Tong J, Zhu WG, Jiang Y, Sun B. Targeting NAT10 attenuates homologous recombination via destabilizing DNA:RNA hybrids and overcomes PARP inhibitor resistance in cancers. Drug Resist Updat 2025; 81:101241. [PMID: 40132530 DOI: 10.1016/j.drup.2025.101241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025]
Abstract
AIMS RNA metabolism has been extensively studied in DNA double-strand break (DSB) repair. The RNA acetyltransferase N-acetyltransferase 10 (NAT10)-mediated N4-acetylcytidine (ac4C) modification in DSB repair remains largely elusive. In this study, we aim to decipher the role for ac4C modification by NAT10 in DSB repair in hepatocellular carcinoma (HCC). METHODS Laser micro-irradiation and chromatin immunoprecipitation (ChIP) were used to assess the accumulation of ac4C modification and NAT10 at DSB sites. Cryo-electron microscopy (cryo-EM) was used to determine the structures of NAT10 in complex with its inhibitor, remodelin. Hepatocyte-specific deletion of NAT10 mouse models were adopted to detect the effects of NAT10 on HCC progression. Subcutaneous xenograft, human HCC organoid and patient-derived xenograft (PDX) model were exploited to determine the therapy efficiency of the combination of a poly (ADP-ribose) polymerase 1 (PARP1) inhibitor (PARPi) and remodelin. RESULTS NAT10 promptly accumulates at DSB sites, where it executes ac4C modification on RNAs at DNA:RNA hybrids dependent on PARP1. This in turn enhances the stability of DNA:RNA hybrids and promotes homologous recombination (HR) repair. The ablation of NAT10 curtails HCC progression. Furthermore, the cryo-EM yields a remarkable 2.9 angstroms resolution structure of NAT10-remodelin, showcasing a C2 symmetric architecture. Remodelin treatment significantly enhanced the sensitivity of HCC cells to a PARPi and targeting NAT10 also restored sensitivity to a PARPi in ovarian and breast cancer cells that had developed resistance. CONCLUSION Our study elucidated the mechanism of NAT10-mediated ac4C modification in DSB repair, revealing that targeting NAT10 confers synthetic lethality to PARP inhibition in HCC. Our findings suggest that co-inhibition of NAT10 and PARP1 is an effective novel therapeutic strategy for patients with HCC and have the potential to overcome PARPi resistance.
Collapse
Affiliation(s)
- Zhu Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China; Department of Cell Biology, School of Life Science, Anhui Medical University, Hefei, Anhui, China
| | - Mingming Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Longpo Geng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Jun Zhang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Jing Xia
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Hongda An
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Anliang Xia
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Yuanyuan Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Shihan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Junjie Tong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China; Department of Cell Biology, School of Life Science, Anhui Medical University, Hefei, Anhui, China
| | - Wei-Guo Zhu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Yiyang Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China; Department of Cell Biology, School of Life Science, Anhui Medical University, Hefei, Anhui, China.
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; MOE Innovation Center for Basic Research in Tumor Immunotherapy, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China.
| |
Collapse
|
13
|
Zhang X, Qin S, Huang F, Liu H, Wang J, Chen Z, Hao H, Ding S, Liu L, Yu B, Liu Y, Liu H, Guan W. N4-acetylcytidine coordinates with NP1 and CPSF5 to facilitate alternative RNA processing during the replication of minute virus of canines. Nucleic Acids Res 2025; 53:gkaf229. [PMID: 40167508 PMCID: PMC11959542 DOI: 10.1093/nar/gkaf229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/07/2025] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
RNA modifications play crucial roles in RNA metabolism, structure, and functions. N4-acetylcytidine (ac4C) modifications have been shown to enhance stability and translation efficiency of messenger RNAs and viral RNAs. However, the relationship between ac4C and alternative RNA processing remains unexplored. Here, N-acetyltransferase 10 (NAT10) and its catalyzed ac4C modifications on minute virus of canines (MVC) were shown to regulate viral DNA replication and RNA processing, including both the alternative RNA splicing and polyadenylation. Through acRIP-seq and RedaC:T-seq, functional ac4C-modified residue 3311 was identified and characterized, which affected MVC RNA processing rather than altered the viral RNA stability. Ac4C modification at nt 3311 was revealed to participate in NP1-mediated viral RNA processing without influencing RNA affinity of NP1. Meanwhile, CPSF5 was identified to interact with NP1 and mediate viral RNA processing in an ac4C-dependent manner. Further in vitro assays showed that NP1 recruited CPSF5 to MVC RNAs, and the ac4C modification promoted specific binding of CPSF5 to the target region, which ensured precise alternative MVC RNA processing. This study not only reveals the functions of NAT10 and ac4C but also elucidates the mechanisms by which RNA modifications orchestrate MVC proteins and host factors for efficient viral replication and alternative RNA processing.
Collapse
Affiliation(s)
- Xueyan Zhang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Shuangkang Qin
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Huang
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Haizhou Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Jun Wang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Zhen Chen
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Haojie Hao
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Shuang Ding
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Lishi Liu
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Baocheng Yu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Liu
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Haibin Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Wuxiang Guan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| |
Collapse
|
14
|
Hu M, Lv L, Lei Y, Chen M, Zhou S, Liu Z. NAT10 mediates TLR2 to promote podocyte senescence in adriamycin-induced nephropathy. Cell Death Dis 2025; 16:185. [PMID: 40108127 PMCID: PMC11923244 DOI: 10.1038/s41419-025-07515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
N-acetyltransferase 10 (NAT10) is involved in regulating senescence. However, its role in glomerular diseases remains unclear. Therefore, this study aims to investigate the mechanisms by which NAT10 influences senescence and damage in an adriamycin (ADR)-induced nephropathy model. Senescence (p16 and p21) and DNA damage markers (γ-H2AX (ser139)) were assessed in ADR-induced nephropathy. NAT10 function was demonstrated using Remodelin or small interfering RNA (siRNA) interventions. Transcriptome sequencing was conducted to identify key downstream genes and pathways, while coimmunoprecipitation was performed to evaluate the relationship between NAT10 and toll-like receptor 2 (TLR2) expression. TLR2 overexpression or knockdown further validated its regulatory role in senescence. In ADR-treated mice, the expression levels of P53, P21, P16, γ-H2AX(S139) proteins were elevated, while those of WT-1 and nephrin were reduced. This effect was mitigated by Remodelin and siNAT10 administration. Transcriptome sequencing identified TLR2 as a key downstream gene, and coimmunoprecipitation, along with molecular docking models, confirmed its interaction with NAT10. TLR2 overexpression plasmid or siRNA was employed for recovery experiments. Together, the study findings suggest that NAT10 contributes to podocyte senescence and injury via interaction with TLR2. Further, it demonstrates that NAT10 alleviates ADR-induced podocyte senescence by interacting with TLR2, potentially through a P53-P21-dependent mechanism. Thus NAT10 could serve as a novel therapeutic target for treating podocyte senescence and proteinuric glomerulopathies.
Collapse
Affiliation(s)
- Mingyang Hu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Linxiao Lv
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Yuqi Lei
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Min Chen
- Institute of Nephrology, Peking University, Beijing, PR China
| | - Sijie Zhou
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China.
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China.
| | - Zhangsuo Liu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China.
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China.
| |
Collapse
|
15
|
Li H, Cai X, Xu C, Yang X, Song X, Kong Y, Yang M, Wu Q, Zheng SG, Shao Y, Wang P, Zhou J, Li HB. RNA cytidine acetyltransferase NAT10 maintains T cell pathogenicity in inflammatory bowel disease. Cell Discov 2025; 11:19. [PMID: 40038243 DOI: 10.1038/s41421-025-00781-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
The emerging field of epitranscriptomics is reshaping our understanding of post-transcriptional gene regulation in inflammatory diseases. N4-acetylcytidine (ac4C), the only known acetylation modification in RNA catalyzed by N-acetyltransferase 10 (NAT10), is known to enhance mRNA stability and translation, yet its role in inflammatory bowel disease (IBD) remains unclear. In this study, we discovered that Nat10 expression correlates with inflammatory and apoptotic pathways in human ulcerative colitis CD4+ T cells. Our further analysis revealed that the deficiency of NAT10 led to a disruption of T cell development at steady state, and identified a pivotal role for NAT10 in preserving the pathogenicity of naïve CD4+ T cells to induce adoptive transfer colitis. Mechanistically, the lack of NAT10 triggers the diminished stability of the anti-apoptotic gene BCL2-associated athanogene 3 (Bag3), initiating a cascade of events that includes the upregulation of apoptosis-related genes and an accelerated rate of apoptosis in T cells. Our findings reveal a previously unrecognized role of the NAT10-ac4C-Bag3 axis in preserving T cell balance and suggests that targeting RNA ac4C modification could be a promising therapeutic approach for IBD.
Collapse
Affiliation(s)
- Haixin Li
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuemin Cai
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| | - Changfen Xu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinhui Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaohan Song
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxin Kong
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Mei Yang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qielan Wu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Song Guo Zheng
- Department of Rheumatology & Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiming Shao
- The Key Laboratory of Sepsis Translational Medicine, Guangdong Medical University; Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Jing Zhou
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hua-Bing Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China.
- Department of Geriatrics, Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
16
|
Schwarz FM, Klotz DM, Yang R, Brux M, Buchholz F, Harb H, Link T, Wimberger P, Theis M, Kuhlmann JD. Methylstat sensitizes ovarian cancer cells to PARP-inhibition by targeting the histone demethylases JMJD1B/C. Cancer Gene Ther 2025; 32:286-296. [PMID: 39915607 PMCID: PMC11946898 DOI: 10.1038/s41417-025-00874-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: 08/30/2024] [Revised: 01/06/2025] [Accepted: 01/29/2025] [Indexed: 03/28/2025]
Abstract
PARP-inhibitors (PARPi) are an integral part of ovarian cancer treatment. However, overcoming acquired PARPi resistance or increasing the benefit of PARPi in patients without homologous recombination deficiency (HRD) remains an unmet clinical need. We sought to identify genetic modulators of PARPi response, guiding pharmacological PARPi sensitization. CRISPR-Cas9 mediated loss-of-function screen with a focused sgRNA library revealed that DNA-demethylases JMJD1B/JMJD1C, targetable by the small inhibitor methylstat, promote PARPi resistance. Methylstat synergistically interacted with olaparib, and (re-)sensitized ovarian cancer cells to PARPi treatment, surpassing the efficacy of common demethylase inhibitors. Genetic knockout of JMJD1B and/or JMJD1C phenocopied the effect of methylstat in an additive manner. Validation studies revealed methylstat to be a universal PARPi-sensitizing drug, effective, regardless of PARPi resistance status or BRCA1 mutational background. Methylstat modulated clonal cancer dynamics by mitigating positive selection of PARPi-resistant or BRCA1-proficient cells under olaparib treatment. Using a model of PARPi-induced cellular toxicity, we showed that methylstat impairs cellular DNA repair, indicated by an increased susceptibility of ovarian cancer cells to olaparib-induced DNA double strand breaks after methylstat exposure. This study proposes the histone demethylase inhibitor methylstat as an epigenetic drug for overcoming PARPi-resistance or for increasing efficacy of PARPi beyond HRD in ovarian cancer patients.
Collapse
Affiliation(s)
- Franziska Maria Schwarz
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases/University Cancer Center (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, Dresden, Germany
| | - Daniel Martin Klotz
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases/University Cancer Center (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, Dresden, Germany
| | - Ruming Yang
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases/University Cancer Center (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, Dresden, Germany
| | - Melanie Brux
- National Center for Tumor Diseases/University Cancer Center (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Frank Buchholz
- National Center for Tumor Diseases/University Cancer Center (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, Dresden, Germany
- Medical Systems Biology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Hani Harb
- Institute for Medical Microbiology and Virology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Theresa Link
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases/University Cancer Center (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, Dresden, Germany
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases/University Cancer Center (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, Dresden, Germany
| | - Mirko Theis
- National Center for Tumor Diseases/University Cancer Center (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Medical Systems Biology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jan Dominik Kuhlmann
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- National Center for Tumor Diseases/University Cancer Center (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, Dresden, Germany.
| |
Collapse
|
17
|
Chen L, Wang WJ, Liu SY, Su RB, Wu YK, Wu X, Zhang SY, Qiao J, Sha QQ, Fan HY. NAT10-mediated mRNA N4-acetylation is essential for the translational regulation during oocyte meiotic maturation in mice. SCIENCE ADVANCES 2025; 11:eadp5163. [PMID: 39982985 PMCID: PMC11844725 DOI: 10.1126/sciadv.adp5163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 01/17/2025] [Indexed: 02/23/2025]
Abstract
The precise translational regulation of maternal messenger RNAs (mRNAs) drives mammalian oocyte maturation. However, the function and mechanism of posttranscriptional chemical modifications, especially the newly identified N4-acetylcytidine (ac4C) modification catalyzed by N-acetyltransferase 10 (NAT10), are unknown. In this study, we developed a low-input ac4C sequencing technology, ac4C LACE-seq, and mapped 8241 ac4C peaks at the whole-transcriptome level using 50 mouse oocytes at the germinal vesicle stage. Oocyte-specific Nat10 knockout wiped out ac4C signals in oocytes and caused severe defects in meiotic maturation and female infertility. Mechanically, Nat10 deletion led to a failure of ac4C deposition on mRNAs encoding key maternal factors, which regulate transcriptome stability and maternal-to-zygotic transition. Nat10-deleted oocytes showed decreased mRNA translation efficiency due to the direct inhibition of ac4C sites on specific transcripts during meiotic maturation. In summary, we developed a low-input, high-sensitivity mRNA ac4C profiling approach and highlighted the important physiological function of ac4C in the precise regulation of oocyte meiotic maturation by enhancing translation efficiency.
Collapse
Affiliation(s)
- Lu Chen
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Wen-Jing Wang
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Shao-Yuan Liu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Rui-Bao Su
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Yu-Ke Wu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Xuan Wu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Song-Ying Zhang
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Qian-Qian Sha
- College of Life Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Heng-Yu Fan
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Center for Biomedical Research, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China
| |
Collapse
|
18
|
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
|
19
|
Breusegem SY, Houghton J, Romero-Bueno R, Fragoso-Luna A, Kentistou KA, Ong KK, Janssen AFJ, Bright NA, Riedel CG, Perry JRB, Askjaer P, Larrieu D. A multiparametric anti-aging CRISPR screen uncovers a role for BAF in protein synthesis regulation. Nat Commun 2025; 16:1681. [PMID: 39956852 PMCID: PMC11830792 DOI: 10.1038/s41467-025-56916-5] [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: 07/31/2023] [Accepted: 01/28/2025] [Indexed: 02/18/2025] Open
Abstract
Progeria syndromes are very rare, incurable premature aging conditions recapitulating most aging features. Here, we report a whole genome, multiparametric CRISPR screen, identifying 43 genes that can rescue multiple cellular phenotypes associated with progeria. We implement the screen in fibroblasts from Néstor-Guillermo Progeria Syndrome male patients, carrying a homozygous A12T mutation in BAF. The hits are enriched for genes involved in protein synthesis, protein and RNA transport and osteoclast formation and are validated in a whole-organism Caenorhabditis elegans model. We further confirm that BAF A12T can disrupt protein synthesis rate and fidelity, which could contribute to premature aging in patients. This work highlights the power of multiparametric genome-wide suppressor screens to identify genes enhancing cellular resilience in premature aging and provide insights into the biology underlying progeria-associated cellular dysfunction.
Collapse
Affiliation(s)
- Sophia Y Breusegem
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Sophia Y. Breusegem: MRC toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Jack Houghton
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Jack Houghton: Imperial College London, Exhibition Road, South Kensington, London, UK
| | - Raquel Romero-Bueno
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Adrián Fragoso-Luna
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Katherine A Kentistou
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Anne F J Janssen
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Anne F. J. Janssen: Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, The Netherlands
| | - Nicholas A Bright
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
| | | | - John R B Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Peter Askjaer
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Delphine Larrieu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK.
- Delphine Larrieu: Altos Labs, Cambridge Institute of Science, Cambridge, UK.
| |
Collapse
|
20
|
Han Y, Zhang X, Miao L, Lin H, Zhuo Z, He J, Fu W. Biological function and mechanism of NAT10 in cancer. CANCER INNOVATION 2025; 4:e154. [PMID: 39817252 PMCID: PMC11732740 DOI: 10.1002/cai2.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 01/18/2025]
Abstract
N-acetyltransferase 10 (NAT10) is a nucleolar acetyltransferase with an acetylation catalytic function and can bind various protein and RNA molecules. As the N4-acetylcytidine (ac4C) "writer" enzyme, NAT10 is reportedly involved in a variety of physiological and pathological activities. Currently, the NAT10-related molecular mechanisms in various cancers are not fully understood. In this review, we first describe the cellular localization of NAT10 and then summarize its numerous biological functions. NAT10 is involved in various biological processes by mediating the acetylation of different proteins and RNAs. These biological functions are also associated with cancer progression and patient prognosis. We also review the mechanisms by which NAT10 plays roles in various cancer types. NAT10 can affect tumor cell proliferation, metastasis, and stress tolerance through its acetyltransferase properties. Further research into NAT10 functions and expression regulation in tumors will help explore its future potential in cancer diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Yufeng Han
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xinxin Zhang
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Lei Miao
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Huiran Lin
- Faculty of MedicineMacau University of Science and TechnologyMacauChina
| | - Zhenjian Zhuo
- Laboratory Animal Center, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenGuangdongChina
- State Key Laboratory of Chemical OncogenomicsPeking University Shenzhen Graduate SchoolShenzhenGuangdongChina
| | - Jing He
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Wen Fu
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of PediatricsGuangzhou Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
21
|
Liu N, YangOu J, Wei C, Li G, Yu R, Lin Y, Xu H. NAT10 drives endometriosis progression through acetylation and stabilization of TGFB1 mRNA. Mol Cell Endocrinol 2025; 597:112447. [PMID: 39732360 DOI: 10.1016/j.mce.2024.112447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
Endometriosis, a gynecological disorder marked by pelvic pain and infertility, has its pathogenesis and pathophysiology significantly influenced by epigenetics, as these factors have been well characterized. However, the role of RNA-mediated epigenetic regulation in endometriosis remains to be elucidated. In our study, we found that N4-acetylcytidine (ac4C) RNA modification and N-acetyltransferase 10 (NAT10) were significantly upregulated in endometrial lesions compared to eutopic endometrium. Knockdown of NAT10 suppressed endometrial epithelial cell proliferation, epithelial-to-mesenchymal transition (EMT), and cell cycle processes in vitro. RNA-seq and acRIP-seq analyses revealed that the knockdown of NAT10 impaired cell proliferation and the TGF-beta signaling pathway. We further identified that ac4C RNA modification enhanced TGFB1 mRNA stability and expression levels, and inhibition of NAT10 activity by Remodelin effectively suppressed the growth of ectopic lesions in an endometriosis mouse model. Collectively, our findings reveal that increased NAT10-mediated ac4C modification enhances TGFB1 mRNA stability, thereby promoting the development of endometriosis. This discovery lays the molecular foundation for future therapeutic approaches targeting endometriosis.
Collapse
Affiliation(s)
- Na Liu
- International Peace Maternity & Child Health Hospital, Shanghai Municipal Key Clinical Speciality, Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jing YangOu
- International Peace Maternity & Child Health Hospital, Shanghai Municipal Key Clinical Speciality, Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Chenxuan Wei
- International Peace Maternity & Child Health Hospital, Shanghai Municipal Key Clinical Speciality, Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Guojing Li
- International Peace Maternity & Child Health Hospital, Shanghai Municipal Key Clinical Speciality, Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Ruoer Yu
- International Peace Maternity & Child Health Hospital, Shanghai Municipal Key Clinical Speciality, Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yu Lin
- International Peace Maternity & Child Health Hospital, Shanghai Municipal Key Clinical Speciality, Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Hong Xu
- International Peace Maternity & Child Health Hospital, Shanghai Municipal Key Clinical Speciality, Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
22
|
Luo R, Zhang Y, Kumata K, Xie L, Kurihara Y, Ogawa M, Kokufuta T, Nengaki N, Wang F, Zhang MRR. The N-acetyltransferase 10 inhibitor [ 11C]remodelin: synthesis and preliminary positron emission tomography study in mice. EJNMMI Radiopharm Chem 2025; 10:6. [PMID: 39888477 PMCID: PMC11785860 DOI: 10.1186/s41181-025-00330-1] [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: 12/26/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND 4-(4-Cyanophenyl)-2-(2-cyclopentylidenehydrazinyl)thiazole (remodelin) is a potent N-acetyltransferase 10 (NAT10) inhibitor. This compound inhibits tumors and weakens tumor resistance to antitumor drugs. Moreover, remodelin has been found to enhance healthspan in an animal model of the human accelerated ageing syndrome. In this study, we synthesized C-11-labelled remodelin ([11C]remodelin) for the first time as a positron emission tomography (PET) probe and assessed its biodistribution in mice using PET. RESULTS [11C]Remodelin was synthesized by the reaction of a boron ester precursor (1) with hydrogen [11C]cyanide, which was prepared from the cyclotron-produced [11C]carbon dioxide via [11C]methane. The decay-corrected radiochemical yield of [11C]remodelin was 6.2 ± 2.3% (n = 20, based on [11C]carbon dioxide) with a synthesis time of 45 min and radiochemical purity of > 90%. A PET study with [11C]remodelin showed high uptake of radioactivity in the heart, liver, and small intestine of mice. The metabolite analysis indicated moderate metabolism of [11C]remodelin in the heart. CONCLUSIONS In the present study, we successfully synthesized [11C]remodelin and assessed its biodistribution of radioactivity in the mouse organs and tissues with PET. We are planning to prepare tumor and inflammatory models in which overexpression of NAT10 is possibly induced and conduct PET imaging for these animal models with [11C]remodelin to elucidate the relationship between NAT10 and diseases.
Collapse
Affiliation(s)
- Rui Luo
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
- Department of Nuclear Medicine, Nanjing First Hospital, Affiliated to Nanjing Medical University, 68 Chanle Road, Nanjing, 210006, China
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Katsushi Kumata
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
| | - Yusuke Kurihara
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
- SHI Accelerator Service, Ltd, 7-1-1 Nishigotanda, Shinagawa-ku, Tokyo, 141-0031, Japan
| | - Masanao Ogawa
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
- SHI Accelerator Service, Ltd, 7-1-1 Nishigotanda, Shinagawa-ku, Tokyo, 141-0031, Japan
| | - Tomomi Kokufuta
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Nobuki Nengaki
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
- SHI Accelerator Service, Ltd, 7-1-1 Nishigotanda, Shinagawa-ku, Tokyo, 141-0031, Japan
| | - Feng Wang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
- Department of Nuclear Medicine, Nanjing First Hospital, Affiliated to Nanjing Medical University, 68 Chanle Road, Nanjing, 210006, China.
| | - Ming-Rong R Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
23
|
Wang Q, Li M, Chen C, Xu L, Fu Y, Xu J, Shu C, Wang B, Wang Z, Chen C, Song T, Wang S. Glucose homeostasis controls N-acetyltransferase 10-mediated ac4C modification of HK2 to drive gastric tumorigenesis. Theranostics 2025; 15:2428-2450. [PMID: 39990211 PMCID: PMC11840738 DOI: 10.7150/thno.104310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/06/2025] [Indexed: 02/25/2025] Open
Abstract
Rationale: Abnormal metabolic states contribute to a variety of diseases, including cancer. RNA modifications have diverse biological functions and are implicated in cancer development, including gastric cancer (GC). However, the direct relationship between glucose homeostasis and 4-acetylcytosine (ac4C) modification in GC remains unclear. Methods: The prognostic value of RNA acetyltransferase NAT10 expression was evaluated in a human GC cohort. Additionally, preoperative PET/CT data from GC patients and Micro-PET/CT imaging of mice were employed to assess the relationship between NAT10 and glucose metabolism. The biological role of NAT10 in GC was investigated through various experiments, including GC xenografts, organoids, and a conditional knockout (cKO) mouse model. The underlying mechanisms were examined using dot blotting, immunofluorescence staining, co-immunoprecipitation, and high-throughput sequencing, among other techniques. Results: Glucose deprivation activates the autophagy-lysosome pathway, leading to the degradation of NAT10 by enhancing its interaction with the sequestosome 1 (SQSTM1)/microtubule-associated protein 1 light chain 3 alpha (LC3) complex, ultimately resulting in a reduction of ac4C modification. Furthermore, the levels of ac4C and NAT10 are elevated in GC tissues and correlate with poor prognosis. A strong correlation exists between NAT10 levels and 18F-FDG uptake in GC patients. Furthermore, NAT10 drives glycolytic metabolism and gastric carcinogenesis in vitro and in vivo. Mechanistically, NAT10 stimulates ac4C modification at the intersection of the coding sequence (CDS) and 3' untranslated region (3'UTR) of hexokinase 2 (HK2) mRNA, enhancing its stability and activating the glycolytic pathway, thereby driving gastric tumorigenesis. Conclusion: Our findings highlight the critical crosstalk between glucose homeostasis and the ac4C epitranscriptome in gastric carcinogenesis. This finding offers a potential strategy of targeting NAT10/HK2 axis for the treatment of GC patients, especially those with highly active glucose metabolism.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
| | - Mengmeng Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Chen Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
| | - Lei Xu
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yao Fu
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiawen Xu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Chuanjun Shu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Bo Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
| | - Zhangding Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
| | - Changyu Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tao Song
- Department of Gastrointestinal Surgery, Xuzhou Central Hospital, Xuzhou, China
| | - Shouyu Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
24
|
Li Z, Meng K, Lan S, Ren Z, Lai Z, Ao X, Liu Z, Xu J, Mo X, Zhang Z. The Role of mRNA Modifications in Bone Diseases. Int J Biol Sci 2025; 21:1065-1080. [PMID: 39897026 PMCID: PMC11781163 DOI: 10.7150/ijbs.104460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025] Open
Abstract
As a type of epigenetic modifications, mRNA modifications regulate the metabolism of mRNAs, thereby influencing gene expression. Previous studies have indicated that dysregulation of mRNA modifications is closely associated with the occurrence and progression of bone diseases (BDs). In this study, we first introduced the dynamic regulatory processes of five major mRNA modifications and their effects on the nucleus export, stability, and translation of mRNAs. We then summarized the mechanisms of mRNA modifications involved in the development of osteoporosis, osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, fractures, osteomyelitis, and osteosarcoma. Finally, we reviewed therapeutic strategies for BDs based on the above mechanisms, focusing on regulating osteoblast and osteoclast differentiation, inhibiting cellular senescence and injury, and alleviating inflammation. This review identified mRNA modifications as potential targets for treating BDs and proposes perspectives on the diversity, targetability, and safety of mRNA-modifying therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jiajia Xu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaoyi Mo
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
25
|
Hervé S, Scelfo A, Bersano Marchisio G, Grison M, Vaidžiulytė K, Dumont M, Angrisani A, Keikhosravi A, Pegoraro G, Deygas M, P F Nader G, Macé AS, Gentili M, Williart A, Manel N, Piel M, Miroshnikova YA, Fachinetti D. Chromosome mis-segregation triggers cell cycle arrest through a mechanosensitive nuclear envelope checkpoint. Nat Cell Biol 2025; 27:73-86. [PMID: 39779939 PMCID: PMC11735390 DOI: 10.1038/s41556-024-01565-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 10/24/2024] [Indexed: 01/11/2025]
Abstract
Errors during cell division lead to aneuploidy, which is associated with genomic instability and cell transformation. In response to aneuploidy, cells activate the tumour suppressor p53 to elicit a surveillance mechanism that halts proliferation and promotes senescence. The molecular sensors that trigger this checkpoint are unclear. Here, using a tunable system of chromosome mis-segregation, we show that mitotic errors trigger nuclear deformation, nuclear softening, and lamin and heterochromatin alterations, leading to rapid p53/p21 activation upon mitotic exit in response to changes in nuclear mechanics. We identify mTORC2 and ATR as nuclear deformation sensors upstream of p53/p21 activation. While triggering mitotic arrest, the chromosome mis-segregation-induced alterations of nuclear envelope mechanics provide a fitness advantage for aneuploid cells by promoting nuclear deformation resilience and enhancing pro-invasive capabilities. Collectively, this work identifies a nuclear mechanical checkpoint triggered by altered chromatin organization that probably plays a critical role in cellular transformation and cancer progression.
Collapse
Affiliation(s)
- Solène Hervé
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Scelfo
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
| | | | - Marine Grison
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
| | - Kotryna Vaidžiulytė
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Marie Dumont
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
| | - Annapaola Angrisani
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
| | - Adib Keikhosravi
- High-Throughput Imaging Facility, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gianluca Pegoraro
- High-Throughput Imaging Facility, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mathieu Deygas
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Guilherme P F Nader
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anne-Sophie Macé
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France
- CNRS UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Institut Curie, PSL Research University, Paris, France
| | - Matteo Gentili
- INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Alice Williart
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Nicolas Manel
- INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Matthieu Piel
- CNRS UMR144, Institut Curie, Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Yekaterina A Miroshnikova
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Daniele Fachinetti
- CNRS UMR144 - UMR3664, Institut Curie, Sorbonne Université, PSL Research University, Paris, France.
| |
Collapse
|
26
|
Soggia G, ElMaghloob Y, Boromangnaeva AK, Al Jord A. Mechanical Remodeling of Nuclear Biomolecular Condensates. Physiology (Bethesda) 2025; 40:0. [PMID: 39109673 DOI: 10.1152/physiol.00027.2024] [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: 06/03/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
Organism health relies on cell proliferation, migration, and differentiation. These universal processes depend on cytoplasmic reorganization driven notably by the cytoskeleton and its force-generating motors. Their activity generates forces that mechanically agitate the cell nucleus and its interior. New evidence from reproductive cell biology revealed that these cytoskeletal forces can be tuned to remodel nuclear membraneless compartments, known as biomolecular condensates, and regulate their RNA processing function for the success of subsequent cell division that is critical for fertility. Both cytoskeletal and nuclear condensate reorganization are common to numerous physiological and pathological contexts, raising the possibility that mechanical remodeling of nuclear condensates may be a much broader mechanism regulating their function. Here, we review this newfound mechanism of condensate remodeling and venture into the contexts of health and disease where it may be relevant, with a focus on reproduction, cancer, and premature aging.
Collapse
Affiliation(s)
- Giulia Soggia
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Yasmin ElMaghloob
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Systems Biology and Immunology Lab, Children's Cancer Hospital Egypt, Cairo, Egypt
| | | | - Adel Al Jord
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
27
|
Macedo JC, da Silva MM, Logarinho E. Misshapen chromosomes in check by mechanics. Nat Cell Biol 2025; 27:9-11. [PMID: 39695243 DOI: 10.1038/s41556-024-01573-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Affiliation(s)
- Joana C Macedo
- Ageing and Aneuploidy Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Maria M da Silva
- Ageing and Aneuploidy Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Elsa Logarinho
- Ageing and Aneuploidy Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal.
| |
Collapse
|
28
|
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
|
29
|
Zhang S, Huang F, Wang Y, Long Y, Li Y, Kang Y, Gao W, Zhang X, Wen Y, Wang Y, Pan L, Xia Y, Yang Z, Yang Y, Mo H, Li B, Hu J, Song Y, Zhang S, Dong S, Du X, Li Y, Liu Y, Liao W, Gao Y, Zhang Y, Chen H, Liang Y, Chen J, Weng H, Huang H. NAT10-mediated mRNA N 4-acetylcytidine reprograms serine metabolism to drive leukaemogenesis and stemness in acute myeloid leukaemia. Nat Cell Biol 2024; 26:2168-2182. [PMID: 39506072 PMCID: PMC11628400 DOI: 10.1038/s41556-024-01548-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/27/2024] [Indexed: 11/08/2024]
Abstract
RNA modification has emerged as an important epigenetic mechanism that controls abnormal metabolism and growth in acute myeloid leukaemia (AML). However, the roles of RNA N4-acetylcytidine (ac4C) modification in AML remain elusive. Here, we report that ac4C and its catalytic enzyme NAT10 drive leukaemogenesis and sustain self-renewal of leukaemic stem cells/leukaemia-initiating cells through reprogramming serine metabolism. Mechanistically, NAT10 facilitates exogenous serine uptake and de novo biosynthesis through ac4C-mediated translation enhancement of the serine transporter SLC1A4 and the transcription regulators HOXA9 and MENIN that activate transcription of serine synthesis pathway genes. We further characterize fludarabine as an inhibitor of NAT10 and demonstrate that pharmacological inhibition of NAT10 targets serine metabolic vulnerability, triggering substantial anti-leukaemia effects both in vitro and in vivo. Collectively, our study demonstrates the functional importance of ac4C and NAT10 in metabolism control and leukaemogenesis, providing insights into the potential of targeting NAT10 for AML therapy.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Humans
- Animals
- Mice
- Serine/metabolism
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/drug effects
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- N-Terminal Acetyltransferases/metabolism
- N-Terminal Acetyltransferases/genetics
- Cell Line, Tumor
- Homeodomain Proteins/metabolism
- Homeodomain Proteins/genetics
- Cytidine/analogs & derivatives
- Cytidine/pharmacology
- Cytidine/metabolism
- Gene Expression Regulation, Leukemic/drug effects
- Mice, Inbred NOD
- Carcinogenesis/genetics
- Carcinogenesis/metabolism
Collapse
Affiliation(s)
- Subo Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Feng Huang
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Yushuai Wang
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Yifei Long
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanpei Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yalin Kang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Weiwei Gao
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiuxin Zhang
- Bioland Laboratory, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yueting Wen
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Yun Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lili Pan
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
- Union Clinical Medical Colleges, Fujian Medical University, Fuzhou, China
| | - Youmei Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhoutian Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying Yang
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Hongjie Mo
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Baiqing Li
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Jiacheng Hu
- Bioland Laboratory, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yunda Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shilin Zhang
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Shenghua Dong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao Du
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yingmin Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yadi Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenting Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yijun Gao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongming Chen
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Yang Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianjun Chen
- Department of Systems Biology & Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
| | - Hengyou Weng
- Guangzhou National Laboratory, The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China.
- Bioland Laboratory, Guangzhou, China.
| | - Huilin Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
30
|
Attar AG, Paturej J, Banigan EJ, Erbaş A. Chromatin phase separation and nuclear shape fluctuations are correlated in a polymer model of the nucleus. Nucleus 2024; 15:2351957. [PMID: 38753956 PMCID: PMC11407394 DOI: 10.1080/19491034.2024.2351957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 05/18/2024] Open
Abstract
Abnormal cell nuclear shapes are hallmarks of diseases, including progeria, muscular dystrophy, and many cancers. Experiments have shown that disruption of heterochromatin and increases in euchromatin lead to nuclear deformations, such as blebs and ruptures. However, the physical mechanisms through which chromatin governs nuclear shape are poorly understood. To investigate how heterochromatin and euchromatin might govern nuclear morphology, we studied chromatin microphase separation in a composite coarse-grained polymer and elastic shell simulation model. By varying chromatin density, heterochromatin composition, and heterochromatin-lamina interactions, we show how the chromatin phase organization may perturb nuclear shape. Increasing chromatin density stabilizes the lamina against large fluctuations. However, increasing heterochromatin levels or heterochromatin-lamina interactions enhances nuclear shape fluctuations by a "wetting"-like interaction. In contrast, fluctuations are insensitive to heterochromatin's internal structure. Our simulations suggest that peripheral heterochromatin accumulation could perturb nuclear morphology, while nuclear shape stabilization likely occurs through mechanisms other than chromatin microphase organization.
Collapse
Affiliation(s)
- Ali Goktug Attar
- UNAM-National Nanotechnology Research Center and Institute of Materials Science & Nanotechnology, Bilkent University, Ankara, Turkey
| | | | - Edward J Banigan
- Institute for Medical Engineering and Science and Department of Physics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aykut Erbaş
- UNAM-National Nanotechnology Research Center and Institute of Materials Science & Nanotechnology, Bilkent University, Ankara, Turkey
- Institute of Physics, University of Silesia, Chorzów, Poland
| |
Collapse
|
31
|
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
|
32
|
Zou Y, Guo Z, Ge XY, Qiu Y. RNA Modifications in Pathogenic Viruses: Existence, Mechanism, and Impacts. Microorganisms 2024; 12:2373. [PMID: 39597761 PMCID: PMC11596894 DOI: 10.3390/microorganisms12112373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
RNA modification is a key posttranscriptional process playing various biological roles, and one which has been reported to exist extensively in cellular RNAs. Interestingly, recent studies have shown that viral RNAs also contain a variety of RNA modifications, which are regulated dynamically by host modification machinery and play critical roles in different stages of the viral life cycle. In this review, we summarize the reports of four typical modifications reported on viral RNAs, including N6-methyladenosine (m6A), 5-methylcytosine (m5C), N4-acetylcytosine (ac4C), and N1-methyladenosine (m1A), describe the molecular mechanisms of these modification processes, and illustrate their impacts on viral replication, pathogenicity, and innate immune responses. Notably, we find that RNA modifications in different viruses share some common features and mechanisms in their generation, regulation, and function, highlighting the potential for viral RNA modifications and the related host machinery to serve as the targets or bases for the development of antiviral therapeutics and vaccines.
Collapse
Affiliation(s)
| | | | - Xing-Yi Ge
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410012, China; (Y.Z.); (Z.G.)
| | - Ye Qiu
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410012, China; (Y.Z.); (Z.G.)
| |
Collapse
|
33
|
Wang Z, Wan X, Khan MA, Peng L, Sun X, Yi X, Chen K. NAT10 promotes liver lipogenesis in mouse through N4-acetylcytidine modification of Srebf1 and Scap mRNA. Lipids Health Dis 2024; 23:368. [PMID: 39529018 PMCID: PMC11552140 DOI: 10.1186/s12944-024-02360-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Metabolic dysfunction associated steatotic liver disease (MASLD), closely linked to excessive lipogenesis, induces chronic liver disease. MASLD often cause other metabolic diseases, such as cardiovascular disease, diabetes and obesity. However, the mechanism of N-acetyltransferase 10 (NAT10)-mediated N4-acetylcytidine (ac4C) mRNA modification in lipogenesis of MASLD has not been fully elucidated. This study investigated the role of NAT10 in lipogenesis targeting mRNA ac4C modification. METHODS The expression of NAT10 in mouse liver was assessed after a 12-week high-fat diet. In addition, the expression of NAT10 also was detected after AML12 hepatocytes cells were treated with 150 µmol/L palmitic acid (PA). The ac4C mRNA modification was performed by dot blotting. Oil red O staining and the mRNA expression of Srebf1, Acaca and Fasn were used to assess lipogenesis in AML12 cells with NAT10 overexpression or knockdown. acRIP-PCR and NAT10 RIP-PCR were used to verify the Srebf1 and Scap mRNA ac4C modification by NAT10. Furthermore, the liver lipogenesis was evaluated by AAV-mediated target knockdown of NAT10 in mouse liver and treating a specific inhibitor, Remodelin. RESULTS This study revealed that NAT10 is significantly upregulated in liver lipogenesis after a 12-week high-fat diet. NAT10 and ac4C mRNA modification were also drastically increased in AML12 cells after treated with 150 µmol/L PA. Silencing of NAT10 notably inhibited the lipogenesis in AML12 cells and AAV-mediated target knockdown of NAT10 in mouse liver. The acRIP-PCR and NAT10-RIP-PCR revealed that NAT10 ac4C modified Srebf1 and Scap mRNA, the critical modulator of liver lipogenesis, to regulate liver lipogenesis. Besides, Remodelin strongly inhibited liver lipogenesis, including liver TG, serum ALT, AST, TG and TC level and glucose metabolism. CONCLUSIONS NAT10 mediates ac4C modification of Srebf1 and Scap mRNA, thereby affecting lipogenesis in the liver. This study provided a new target for the treatment of MASLD.
Collapse
Affiliation(s)
- Zhouqi Wang
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha, Hunan, 410013, People's Republic of China
| | - Xinxing Wan
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha, Hunan, 410013, People's Republic of China
| | - Md Asaduzzaman Khan
- Department of Biochemistry and Microbiology, School of Health & Life Sciences, North South University, Plot-15, Block-B, Bashundhara, Dhaka, 1229, Bangladesh
| | - Lin Peng
- Department of Nephrology, The First Hospital of Changsha, No. 311 Yingpan Road, Kaifu District, Changsha, Hunan, 410005, People's Republic of China
| | - Xiaoying Sun
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha, Hunan, 410013, People's Republic of China
| | - Xuan Yi
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha, Hunan, 410013, People's Republic of China
| | - Ke Chen
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha, Hunan, 410013, People's Republic of China.
| |
Collapse
|
34
|
Li J, Yushanjiang F, Fang Z, Liu W. NAT10-mediated RNA ac4C acetylation contributes to the myocardial infarction-induced cardiac fibrosis. J Cell Mol Med 2024; 28:e70141. [PMID: 39482983 PMCID: PMC11528131 DOI: 10.1111/jcmm.70141] [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: 09/22/2023] [Revised: 08/31/2024] [Accepted: 10/01/2024] [Indexed: 11/03/2024] Open
Abstract
Cardiac fibrosis is featured cardiac fibroblast activation and extracellular matrix accumulation. Ac4C acetylation is an important epigenetic regulation of RNAs that has been recently discovered, and it is solely carried out by NAT10, the exclusive enzyme used for the modification. However, the potential regulatory mechanisms of ac4C acetylation in myocardial fibrosis following myocardial infarction remain poorly understood. In our study, we activated fibroblasts in vitro using TGF-β1 (20 ng/mL), followed by establishing a myocardial infarction mouse model to evaluate the impact of NAT10 on collagen synthesis and cardiac fibroblast proliferation. We utilized a NAT10 inhibitor, Remodelin, to attenuate the acetylation capacity of NAT10. In the cardiac fibrosis tissues of chronic myocardial infarction mice and cultured cardiac fibroblasts (CFs) in response to TGF-β1 treatment, there was an elevation in the levels of NAT10 expression. This increase facilitated proliferation, the accumulation of collagens, as well as fibroblast-to-myofibroblast transition. Through the administration of Remodelin, we effectively reduced cardiac fibrosis in myocardial infarction mice by inhibiting NAT10's ability to acetylate mRNA. Inhibition of NAT10 resulted in changes in collagen-related gene expression and ac4C acetylation levels. Mechanistically, we found that NAT10 upregulates the acetylation modification of BCL-XL mRNA and enhances the stability of BCL-XL mRNA, thereby upregulating its protein expression, inhibiting the activation of Caspase3 and blocking the apoptosis of CFs. Therefore, the crucial involvement of NAT10-mediated ac4C acetylation is significant in the cardiac fibrosis progression, affording promising molecular targets for the treatment of fibrosis and relevant cardiac diseases.
Collapse
Affiliation(s)
- Jun Li
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Cardiovascular Research InstituteWuhan UniversityWuhanHubeiChina
- Hubei Key Laboratory of CardiologyWuhanHubeiChina
| | - Feierkaiti Yushanjiang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Cardiovascular Research InstituteWuhan UniversityWuhanHubeiChina
- Hubei Key Laboratory of CardiologyWuhanHubeiChina
| | - Zhao Fang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Cardiovascular Research InstituteWuhan UniversityWuhanHubeiChina
- Hubei Key Laboratory of CardiologyWuhanHubeiChina
| | - Wan‐li Liu
- Department of Pediatric, Maternal and Child Health Hospital of Hubei Province, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
35
|
Li WP, Mao XT, Xie JH, Li JY, Liu BQ, Wu LX, Yang B, Li YY, Jin J. N-acetyltransferase 10 is implicated in the pathogenesis of cycling T cell-mediated autoimmune and inflammatory disorders in mice. Nat Commun 2024; 15:9388. [PMID: 39477944 PMCID: PMC11525920 DOI: 10.1038/s41467-024-53350-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024] Open
Abstract
T cell expansion has a crucial function in both autoimmune and chronic inflammatory diseases, with cycling T cells contributing to the pathogenesis of autoimmune diseases by causing uncontrolled immune responses and tissue damage. Yet the regulatory mechanisms governing T cell expansion remain incompletely understood. Here we show that the enzyme N-acetyltransferase 10 (NAT10) regulates T cell activation and proliferation upon antigen stimulation. T cell-specific NAT10 deficiency in mice reduces the number of mature T cells in peripheral lymphoid organs. Mechanistically, NAT10 acetylates RACK1 at K185, preventing subsequent RACK1 K48-linked ubiquitination and degradation. The increased RACK1 stability alters ribosome formation and cellular metabolism, leading to enhanced supply of energy and biosynthetic precursors and, eventually, T cell proliferation. Our findings thus highlight the essential function of NAT10 in T cell self-renewal and metabolism and elucidate NAT10 mode of action for the potential development of novel therapies for immune-related disorders.
Collapse
Affiliation(s)
- Wen-Ping Li
- Center for Neuroimmunology and Health Longevity, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xin-Tao Mao
- Center for Neuroimmunology and Health Longevity, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jia-Huan Xie
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jie-Yu Li
- Center for Neuroimmunology and Health Longevity, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Bao-Qin Liu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Le-Xi Wu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, 310016, China
| | - Bing Yang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yi-Yuan Li
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Jin Jin
- Center for Neuroimmunology and Health Longevity, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
36
|
Xu T, Du T, Zhuang X, He X, Yan Y, Wu J, Zhou H, Li Y, Liao X, He J, Liu C, Dong Y, Ou J, Lin S, Chen D, Huang Z. Loss of NAT10 Reduces the Translation of Kmt5a mRNA Through ac4C Modification in Cardiomyocytes and Induces Heart Failure. J Am Heart Assoc 2024; 13:e035714. [PMID: 39392166 PMCID: PMC11935563 DOI: 10.1161/jaha.124.035714] [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: 03/24/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND In the past decade, the biological functions of various RNA modifications in mammals have been uncovered. N4-acetylcytidine (ac4C), a highly conserved RNA modification, has been implicated in human diseases. Despite this, the involvement of RNA ac4C modification in cardiac physiology and pathology remains incompletely understood. NAT10 (N-acetyltransferase 10) stands as the sole acetyltransferase known to catalyze RNA ac4C modification. This study aims to explore the role of NAT10 and ac4C modification in cardiac physiology and pathology. METHODS AND RESULTS Cardiac-specific knockout of NAT10, leading to reduced RNA ac4C modification, during both neonatal and adult stages resulted in severe heart failure. NAT10 deficiency induced cardiomyocyte apoptosis, a crucial step in heart failure pathogenesis, supported by in vitro data. Activation of the p53 signaling pathway was closely associated with enhanced apoptosis in NAT10-deficient cardiomyocytes. As ac4C modification on mRNA influences translational efficiency, we employed ribosome footprints coupled with RNA sequencing to explore genome-wide translational efficiency changes caused by NAT10 deficiency. We identified and validated that the translational efficiency of Kmt5a was suppressed in NAT10 knockout hearts due to reduced ac4C modification on its mRNA. This finding was consistent with the observation that Kmt5a protein levels were reduced in heart failure despite unchanged mRNA expression. Knockdown of Kmt5a in cardiomyocytes recapitulated the phenotype of NAT10 deficiency, including increased cardiomyocyte apoptosis and activated p53 signaling. Finally, overexpression of Kmt5a rescued cardiomyocyte apoptosis and p53 activation induced by NAT10 inhibition. CONCLUSIONS Our study highlights the significance of NAT10 in cardiomyocyte physiology, demonstrating that NAT10 loss is sufficient to induce cardiomyocyte apoptosis and heart failure. NAT10 regulates the translational efficiency of Kmt5a, a key mediator, through mRNA ac4C modification during heart failure.
Collapse
Affiliation(s)
- Ting Xu
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Tailai Du
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Xiaodong Zhuang
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Xin He
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Youchen Yan
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Jialing Wu
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Huimin Zhou
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Yan Li
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
- Division of Cardiac SurgeryNational‐Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Assisted Circulation and Vascular DiseasesChinese Academy of Medical Sciences, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Xinxue Liao
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Jiangui He
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Chen Liu
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
- Key Laboratory of Assisted Circulation and Vascular DiseasesChinese Academy of Medical Sciences, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Yugang Dong
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
| | - Jingsong Ou
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
- Division of Cardiac SurgeryNational‐Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Assisted Circulation and Vascular DiseasesChinese Academy of Medical Sciences, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Shuibin Lin
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Demeng Chen
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Zhan‐Peng Huang
- Department of Cardiology, Center for Translational MedicineInstitute of Precision Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat‐sen University)GuangzhouChina
- Division of Cardiac SurgeryNational‐Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Assisted Circulation and Vascular DiseasesChinese Academy of Medical Sciences, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
37
|
Su K, Zhao Z, Wang Y, Sun S, Liu X, Zhang C, Jiang Y, Du X. NAT10 resolves harmful nucleolar R-loops depending on its helicase domain and acetylation of DDX21. Cell Commun Signal 2024; 22:490. [PMID: 39394182 PMCID: PMC11468200 DOI: 10.1186/s12964-024-01869-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Aberrant accumulation of R-loops leads to DNA damage, genome instability and even cell death. Therefore, the timely removal of harmful R-loops is essential for the maintenance of genome integrity. Nucleolar R-loops occupy up to 50% of cellular R-loops due to the frequent activation of Pol I transcription. However, the mechanisms involved in the nucleolar R-loop resolution remain elusive. The nucleolar acetyltransferase NAT10 harbors a putative RecD helicase domain (RHD), however, if NAT10 acts in the R-loop resolution is still unknown. METHODS NAT10 knockdown cell lines were constructed using CRISPR/Cas9 technology and short hairpin RNA targeting NAT10 mRNA, respectively. The level of R-loops was detected by immunofluorescent staining combined with RNase H treatment. The helicase activity of NAT10 or DDX21 was determined by in vitro helicase experiment. The interaction between NAT10 and DDX21 was verified by co-immunoprecipitation, immunofluorescent staining and GST pull-down experiments. Acetylation sites of DDX21 by NAT10 were analyzed by mass spectrometry. NAT10 knockdown-induced DNA damage was evaluated by immunofluorescent staining and Western blot detecting γH2AX. RESULTS Depletion of NAT10 led to the accumulation of nucleolar R-loops. NAT10 resolves R-loops through an RHD in vitro and in cells. However, Flag-NAT10 ∆RHD mutant still partially reduced R-loop levels in the NAT10-depleted cells, suggesting that NAT10 might resolve R-loops through additional pathways. Further, the acetyltransferase activity of NAT10 is required for the nucleolar R-loop resolution. NAT10 acetylates DDX21 at K236 and K573 to enhance the helicase activity of DDX21 to unwind nucleolar R-loops. The helicase activity of DDX21 significantly decreased by Flag-DDX21 2KR and increased by Flag-DDX21 2KQ in cells and in vitro. Consequently, NAT10 depletion-induced nucleolar R-loop accumulation led to DNA damage, which was rescued by co-expression of Flag-DDX21 2KQ and Flag-NAT10 G641E, demonstrating that NAT10 resolves nucleolar R-loops through bipartite pathways. CONCLUSION We demonstrate that NAT10 is a novel R-loop resolvase and it resolves nucleolar R-loops depending on its helicase activity and acetylation of DDX21. The cooperation of NAT10 and DDX21 provides comprehensive insights into the nucleolar R-loop resolution for maintaining genome stability.
Collapse
Affiliation(s)
- Kunqi Su
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zhuochen Zhao
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yuying Wang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Shiqi Sun
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xiaofeng Liu
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Chunfeng Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yang Jiang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Xiaojuan Du
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
38
|
Zou Y, Guo S, Wen L, Lv D, Tu J, Liao Y, Chen W, Chen Z, Li H, Chen J, Shen J, Xie X. Targeting NAT10 inhibits osteosarcoma progression via ATF4/ASNS-mediated asparagine biosynthesis. Cell Rep Med 2024; 5:101728. [PMID: 39293390 PMCID: PMC11525028 DOI: 10.1016/j.xcrm.2024.101728] [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: 08/08/2023] [Revised: 01/17/2024] [Accepted: 08/15/2024] [Indexed: 09/20/2024]
Abstract
Despite advances in treatment, the prognosis of patients with osteosarcoma remains unsatisfactory, and searching for potential targets is imperative. Here, we identify N4-acetylcytidine (ac4C) acetyltransferase 10 (NAT10) as a candidate therapeutic target in osteosarcoma through functional screening. NAT10 overexpression is correlated with a poor prognosis, and NAT10 knockout inhibits osteosarcoma progression. Mechanistically, NAT10 enhances mRNA stability of activating transcription factor 4 (ATF4) through ac4C modification. ATF4 induces the transcription of asparagine synthetase (ASNS), which catalyzes asparagine (Asn) biosynthesis, facilitating osteosarcoma progression. Utilizing virtual screening, we identify paliperidone and AG-401 as potential NAT10 inhibitors, and both inhibitors are found to bind to NAT10 proteins. Inhibiting NAT10 suppresses osteosarcoma progression in vivo. Combined treatment using paliperidone and AG-401 produces synergistic inhibition for osteosarcoma in patient-derived xenograft (PDX) models. Our findings demonstrate that NAT10 facilitates osteosarcoma progression through the ATF4/ASNS/Asn axis, and pharmacological inhibition of NAT10 may be a feasible therapeutic approach for osteosarcoma.
Collapse
Affiliation(s)
- Yutong Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong, China
| | - Siyao Guo
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lili Wen
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Dongming Lv
- Department of Burns, Wound Repair and Reconstruction, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian Tu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong, China
| | - Yan Liao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong, China
| | - Weidong Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong, China
| | - Ziyun Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong, China
| | - Hongbo Li
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong, China
| | - Junkai Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong, China
| | - Jingnan Shen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong, China.
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong, China.
| |
Collapse
|
39
|
Shokrollahi M, Stanic M, Hundal A, Chan JNY, Urman D, Jordan CA, Hakem A, Espin R, Hao J, Krishnan R, Maass PG, Dickson BC, Hande MP, Pujana MA, Hakem R, Mekhail K. DNA double-strand break-capturing nuclear envelope tubules drive DNA repair. Nat Struct Mol Biol 2024; 31:1319-1330. [PMID: 38632359 DOI: 10.1038/s41594-024-01286-7] [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: 03/09/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024]
Abstract
Current models suggest that DNA double-strand breaks (DSBs) can move to the nuclear periphery for repair. It is unclear to what extent human DSBs display such repositioning. Here we show that the human nuclear envelope localizes to DSBs in a manner depending on DNA damage response (DDR) kinases and cytoplasmic microtubules acetylated by α-tubulin acetyltransferase-1 (ATAT1). These factors collaborate with the linker of nucleoskeleton and cytoskeleton complex (LINC), nuclear pore complex (NPC) protein NUP153, nuclear lamina and kinesins KIF5B and KIF13B to generate DSB-capturing nuclear envelope tubules (dsbNETs). dsbNETs are partly supported by nuclear actin filaments and the circadian factor PER1 and reversed by kinesin KIFC3. Although dsbNETs promote repair and survival, they are also co-opted during poly(ADP-ribose) polymerase (PARP) inhibition to restrain BRCA1-deficient breast cancer cells and are hyper-induced in cells expressing the aging-linked lamin A mutant progerin. In summary, our results advance understanding of nuclear structure-function relationships, uncover a nuclear-cytoplasmic DDR and identify dsbNETs as critical factors in genome organization and stability.
Collapse
Affiliation(s)
- Mitra Shokrollahi
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mia Stanic
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anisha Hundal
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Research Centre, University Health Network, Toronto, Ontario, Canada
| | - Janet N Y Chan
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Defne Urman
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Chris A Jordan
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anne Hakem
- Princess Margaret Cancer Research Centre, University Health Network, Toronto, Ontario, Canada
| | - Roderic Espin
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Jun Hao
- Princess Margaret Cancer Research Centre, University Health Network, Toronto, Ontario, Canada
| | - Rehna Krishnan
- Princess Margaret Cancer Research Centre, University Health Network, Toronto, Ontario, Canada
| | - Philipp G Maass
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brendan C Dickson
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Manoor P Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Miquel A Pujana
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Razqallah Hakem
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Princess Margaret Cancer Research Centre, University Health Network, Toronto, Ontario, Canada.
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Temerty Centre for AI Research and Education in Medicine, University of Toronto, Toronto, Ontario, Canada.
- College of New Scholars, Artists and Scientists, Royal Society of Canada, Ottawa, Ontario, Canada.
| |
Collapse
|
40
|
Gong S, Qiao H, Wang JY, Huang SY, He SW, Zhao Y, Tan XR, Ye ML, Li JY, Liang YL, Huang SW, Chen J, Zhu XH, Liu N, Li YQ. Ac4C modification of lncRNA SIMALR promotes nasopharyngeal carcinoma progression through activating eEF1A2 to facilitate ITGB4/ITGA6 translation. Oncogene 2024; 43:2868-2884. [PMID: 39154122 DOI: 10.1038/s41388-024-03133-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
The dysregulation of long non-coding RNAs (lncRNAs) are involved in regulating tumor progression in multiple manner. However, little is known about whether lncRNA is involved in the translation regulation of proteins. Here, we identified that the suppressor of inflammatory macrophage apoptosis lncRNA (SIMALR) was highly expressed in nasopharyngeal carcinoma (NPC) tissues by analyzing the lncRNA microarray. Clinically, the high expression of SIMALR served as an independent predictor for inferior prognosis in NPC patients. SIMALR functioned as an oncogenic lncRNA that promoted the proliferation and metastasis of NPC cells in vitro and in vivo. Mechanistically, SIMALR served as a critical accelerator of protein synthesis by binding to eEF1A2 (eukaryotic translation elongation factor 1 alpha 2), one of the most crucial regulators in the translation machinery of the eukaryotic cells, and enhancing its endogenous GTPase activity. Furthermore, SIMALR mediated the activation of eEF1A2 phosphorylation to accelerate the translation of ITGB4/ITGA6, ultimately promoting the malignant phenotype of NPC cells. In addition, N-acetyltransferase 10 (NAT10) enhanced the stability of SIMALR and caused its overexpression in NPC through the N4-acetylcytidine (ac4C) modification. In sum, our results illustrate SIMALR functions as an accelerator for protein translation and highlight the oncogenic role of NAT10-SIMALR-eEF1A2-ITGB4/6 axis in NPC.
Collapse
Affiliation(s)
- Sha Gong
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Han Qiao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jing-Yun Wang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Sheng-Yan Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Shi-Wei He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yin Zhao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Xi-Rong Tan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Ming-Liang Ye
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jun-Yan Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Ye-Lin Liang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Sai-Wei Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jun Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Xun-Hua Zhu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Na Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Ying-Qing Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| |
Collapse
|
41
|
Liu D, Kuang Y, Chen S, Li R, Su F, Zhang S, Qiu Q, Lin S, Shen C, Liu Y, Liang L, Wang J, Xu H, Xiao Y. NAT10 promotes synovial aggression by increasing the stability and translation of N4-acetylated PTX3 mRNA in rheumatoid arthritis. Ann Rheum Dis 2024; 83:1118-1131. [PMID: 38724075 DOI: 10.1136/ard-2023-225343] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/22/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE Recent studies indicate that N-acetyltransferase 10 (NAT10)-mediated ac4C modification plays unique roles in tumour metastasis and immune infiltration. This study aimed to uncover the role of NAT10-mediated ac4C in fibroblast-like synoviocytes (FLSs) functions and synovial immune cell infiltration in rheumatoid arthritis (RA). METHODS FLSs were obtained from active established patients with RA. Protein expression was determined by western blotting or immunohistochemistry or multiplexed immunohistochemistry. Cell migration was measured using a Boyden chamber. ac4C-RIP-seq combined with RNA-seq was performed to identify potential targets of NAT10. RNA immunoprecipitation was used to validate the interaction between protein and mRNA. NAT10 haploinsufficiency, inhibitor remodelin or intra-articular Adv-NAT10 was used to suppress arthritis in mice with delayed-type hypersensitivity arthritis (DYHA) and collagen II-induced arthritis (CIA) and rats with CIA. RESULTS We found elevated levels of NAT10 and ac4C in FLSs and synovium from patients with RA. NAT10 knockdown or specific inhibitor treatment reduced the migration and invasion of RA FLSs. Increased NAT10 level in the synovium was positively correlated with synovial infiltration of multiple types of immune cells. NAT10 inhibition in vivo attenuated the severity of arthritis in mice with CIA and DTHA, and rats with CIA. Mechanistically, we explored that NAT10 regulated RA FLS functions by promoting stability and translation efficiency of N4-acetylated PTX3 mRNA. PTX3 also regulated RA FLS aggression and is associated with synovial immune cell infiltration. CONCLUSION Our findings uncover the important roles of NAT10-mediated ac4C modification in promoting rheumatoid synovial aggression and inflammation, indicating that NAT10 may be a potential target for the treatment of RA, even other dysregulated FLSs-associated disorders.
Collapse
Affiliation(s)
- Di Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu Kuang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Simin Chen
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ruiru Li
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fan Su
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuoyang Zhang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qian Qiu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chuyu Shen
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingli Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liuqin Liang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingnan Wang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hanshi Xu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Youjun Xiao
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
42
|
Li K, Hong Y, Yu Y, Xie Z, Lv D, Wang C, Xie T, Chen H, Chen Z, Zeng J, Zhao S. NAT10 Promotes Prostate Cancer Growth and Metastasis by Acetylating mRNAs of HMGA1 and KRT8. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310131. [PMID: 38922788 PMCID: PMC11348116 DOI: 10.1002/advs.202310131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/22/2024] [Indexed: 06/28/2024]
Abstract
N4-acetylcytidine (ac4C) is essential for the development and migration of tumor cells. According to earlier research, N-acetyltransferase 10 (NAT10) can increase messenger RNAs (mRNAs) stability by catalyzing the synthesis of ac4C. However, little is known about NAT10 expression and its role in the acetylation modifications in prostate cancer (PCa). Thus, the biological function of NAT10 in PCa is investigated in this study. Compared to paraneoplastic tissues, the expression of NAT10 is significantly higher in PCa. The NAT10 expression is strongly correlated with the pathological grade, clinical stage, Gleason score, T-stage, and N-stage of PCa. NAT10 has the ability to advance the cell cycle and the epithelial-mesenchymal transition (EMT), both of which raise the malignancy of tumor cells. Mechanistically, NAT10 enhance the stability of high mobility group AT-hook 1 (HMGA1) by acetylating its mRNA, thereby promoting cell cycle progression to improve cell proliferation. In addition, NAT10 improve the stability of Keratin 8 (KRT8) by acetylating its mRNA, which promotes the progression of EMT to improve cell migration. This findings provide a potential prognostic or therapeutic target for PCa.
Collapse
Affiliation(s)
- Kang‐Jing Li
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
- Department of UrologyAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People's HospitalQingyuan511518China
| | - Yaying Hong
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Yu‐Zhong Yu
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Zhiyue Xie
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Dao‐Jun Lv
- Department of UrologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Chong Wang
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Tao Xie
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Hong Chen
- Luoyang Key Laboratory of Organic Functional MoleculesCollege of Food and DrugLuoyang Normal UniversityLuoyangHenan471934P. R. China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNY11439USA
| | - Jianwen Zeng
- Department of UrologyAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People's HospitalQingyuan511518China
| | - Shan‐Chao Zhao
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
- Department of UrologyThe Fifth Affiliated HospitalSouthern Medical UniversityGuangzhou510900China
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhou510500China
| |
Collapse
|
43
|
Wang Y, Su K, Wang C, Deng T, Liu X, Sun S, Jiang Y, Zhang C, Xing B, Du X. Chemotherapy-induced acetylation of ACLY by NAT10 promotes its nuclear accumulation and acetyl-CoA production to drive chemoresistance in hepatocellular carcinoma. Cell Death Dis 2024; 15:545. [PMID: 39085201 PMCID: PMC11291975 DOI: 10.1038/s41419-024-06951-9] [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: 03/21/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Chemotherapeutic efficacy is seriously impeded by chemoresistance in more than half of hepatocellular carcinoma (HCC) patients. However, the mechanisms involved in chemotherapy-induced upregulation of chemoresistant genes are not fully understood. Here, this study unravels a novel mechanism controlling nuclear acetyl-CoA production to activate the transcription of chemoresistant genes in HCC. NAT10 is upregulated in HCC tissues and its upregulation is correlated with poor prognosis of HCC patients. NAT10 is also upregulated in chemoresistant HCC cells. Targeting NAT10 increases the cytotoxicity of chemotherapy in HCC cells and mouse xenografts. Upon chemotherapy, NAT10 translocates from the nucleolus to the nucleus to activate the transcription of CYP2C9 and PIK3R1. Additionally, nuclear acetyl-CoA is specifically upregulated by NAT10. Mechanistically, NAT10 binds with ACLY in the nucleus and acetylates ACLY at K468 to counteract the SQSTM1-mediated degradation upon chemotherapy. ACLY K468-Ac specifically accumulates in the nucleus and increases nuclear acetyl-CoA production to activate the transcription of CYP2C9 and PIK3R1 through enhancing H3K27ac. Importantly, K468 is required for nuclear localization of ACLY. Significantly, ACLY K468-Ac is upregulated in HCC tissues, and ablation of ACLY K468-Ac sensitizes HCC cells and mouse xenografts to chemotherapy. Collectively, these findings identify NAT10 as a novel chemoresistant driver and the blockage of NAT10-mediated ACLY K468-Ac possesses the potential to attenuate HCC chemoresistance.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Acetyl Coenzyme A/metabolism
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Animals
- Acetylation
- Mice
- Cell Nucleus/metabolism
- Cell Line, Tumor
- Mice, Nude
- Coenzyme A Ligases/metabolism
- Coenzyme A Ligases/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- N-Terminal Acetyltransferases/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Mice, Inbred BALB C
- Male
Collapse
Affiliation(s)
- Yuying Wang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Kunqi Su
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chang Wang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Tao Deng
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaofeng Liu
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Shiqi Sun
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yang Jiang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chunfeng Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Baocai Xing
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China.
| | - Xiaojuan Du
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
44
|
Wang M, Zhang J, Qiu J, Ma X, Xu C, Wu Q, Xing S, Chen X, Liu B. Doxycycline decelerates aging in progeria mice. Aging Cell 2024; 23:e14188. [PMID: 38686927 PMCID: PMC11258430 DOI: 10.1111/acel.14188] [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: 12/16/2023] [Revised: 03/26/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Beyond the antimicrobial activity, doxycycline (DOX) exhibits longevity-promoting effect in nematodes, while its effect on mammals is unclear. Here, we applied a mouse model of Hutchinson-Gilford progeria syndrome (HGPS), Zmpste24 knockout (KO) mice, and analyzed the antiaging effect of DOX. We found that the DOX treatment prolongs lifespan and ameliorates progeroid features of Zmpste24 KO mice, including the decline of body and tissue weight, exercise capacity and cortical bone density, and the shortened colon length. DOX treatment alleviates the abnormal nuclear envelope in multiple tissues, and attenuates cellular senescence and cell death of Zmpste24 KO and HGPS fibroblasts. DOX downregulates the level of proinflammatory IL6 in both serum and tissues. Moreover, the elevated α-tubulin (K40) acetylation mediated by NAT10 in progeria, is rescued by DOX treatment in the aorta tissues in Zmpste24 KO mice and fibroblasts. Collectively, our study uncovers that DOX can decelerate aging in progeria mice via counteracting IL6 expression and NAT10-mediated acetylation of α-tubulin.
Collapse
Affiliation(s)
- Ming Wang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Jie Zhang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Jiangping Qiu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Xuan Ma
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Chenzhong Xu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Qiuhuan Wu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Shaojun Xing
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| |
Collapse
|
45
|
Jin C, Gao J, Zhu J, Ao Y, Shi B, Li X. Exosomal NAT10 from esophageal squamous cell carcinoma cells modulates macrophage lipid metabolism and polarization through ac4C modification of FASN. Transl Oncol 2024; 45:101934. [PMID: 38692194 PMCID: PMC11070927 DOI: 10.1016/j.tranon.2024.101934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/06/2024] [Accepted: 03/08/2024] [Indexed: 05/03/2024] Open
Abstract
N-acetyltransferase 10 (NAT10) is acknowledged as a tumor promoter in various cancers due to its role as a regulator of acetylation modification. Tumor-associated macrophages (TAMs) play a pivotal role in the tumor microenvironment (TME). However, the intercellular communication between esophageal squamous cell carcinoma (ESCC) cells and TAMs involving NAT10 remains poorly understood. This study aimed to elucidate the regulatory mechanism of NAT10 in modulating macrophage lipid metabolism and polarization. Experimental evidence was derived from in vitro and in vivo analyses. We explored the association between upregulated NAT10 in ESCC tissues, macrophage polarization, and the therapeutic efficacy of PD-1. Furthermore, we investigated the impact of methyltransferase 3 (METTL3)-induced m6A modification on the increased expression of NAT10 in ESCC cells. Additionally, we examined the role of exosomal NAT10 in stabilizing the expression of fatty acid synthase (FASN) and promoting macrophage M2 polarization through mediating the ac4C modification of FASN. Results indicated that NAT10, packaged by exosomes derived from ESCC cells, promotes macrophage M2 polarization by facilitating lipid metabolism. In vivo animal studies demonstrated that targeting NAT10 could enhance the therapeutic effect of PD-1 on ESCC by mediating macrophage reprogramming. Our findings offer novel insights into improving ESCC treatment through NAT10 targeting.
Collapse
Affiliation(s)
- Chun Jin
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), No.168 Changhai Road, Yangpu District, Shanghai, China
| | - Jian Gao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ji Zhu
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), No.168 Changhai Road, Yangpu District, Shanghai, China
| | - Yongqiang Ao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bowen Shi
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), No.168 Changhai Road, Yangpu District, Shanghai, China.
| | - Xin Li
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), No.168 Changhai Road, Yangpu District, Shanghai, China.
| |
Collapse
|
46
|
Ngubo M, Chen Z, McDonald D, Karimpour R, Shrestha A, Yockell‐Lelièvre J, Laurent A, Besong OTO, Tsai EC, Dilworth FJ, Hendzel MJ, Stanford WL. Progeria-based vascular model identifies networks associated with cardiovascular aging and disease. Aging Cell 2024; 23:e14150. [PMID: 38576084 PMCID: PMC11258467 DOI: 10.1111/acel.14150] [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: 07/12/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Hutchinson-Gilford Progeria syndrome (HGPS) is a lethal premature aging disorder caused by a de novo heterozygous mutation that leads to the accumulation of a splicing isoform of Lamin A termed progerin. Progerin expression deregulates the organization of the nuclear lamina and the epigenetic landscape. Progerin has also been observed to accumulate at low levels during normal aging in cardiovascular cells of adults that do not carry genetic mutations linked with HGPS. Therefore, the molecular mechanisms that lead to vascular dysfunction in HGPS may also play a role in vascular aging-associated diseases, such as myocardial infarction and stroke. Here, we show that HGPS patient-derived vascular smooth muscle cells (VSMCs) recapitulate HGPS molecular hallmarks. Transcriptional profiling revealed cardiovascular disease remodeling and reactive oxidative stress response activation in HGPS VSMCs. Proteomic analyses identified abnormal acetylation programs in HGPS VSMC replication fork complexes, resulting in reduced H4K16 acetylation. Analysis of acetylation kinetics revealed both upregulation of K16 deacetylation and downregulation of K16 acetylation. This correlates with abnormal accumulation of error-prone nonhomologous end joining (NHEJ) repair proteins on newly replicated chromatin. The knockdown of the histone acetyltransferase MOF recapitulates preferential engagement of NHEJ repair activity in control VSMCs. Additionally, we find that primary donor-derived coronary artery vascular smooth muscle cells from aged individuals show similar defects to HGPS VSMCs, including loss of H4K16 acetylation. Altogether, we provide insight into the molecular mechanisms underlying vascular complications associated with HGPS patients and normative aging.
Collapse
Affiliation(s)
- Mzwanele Ngubo
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
| | - Zhaoyi Chen
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Darin McDonald
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Rana Karimpour
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Amit Shrestha
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Julien Yockell‐Lelièvre
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Aurélie Laurent
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Université de StrasbourgStrasbourgFrance
| | - Ojong Tabi Ojong Besong
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- School of BioscienceUniversity of SkövdeSkövdeSweden
| | - Eve C. Tsai
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
- Division of Neurosurgery, Department of Surgery, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - F. Jeffrey Dilworth
- Department of Cell and Regenerative BiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Michael J. Hendzel
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - William L. Stanford
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Department of Biochemistry, Microbiology & ImmunologyUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
47
|
Ding M, Yu Z, Lu T, Hu S, Zhou X, Wang X. N-acetyltransferase 10 facilitates tumorigenesis of diffuse large B-cell lymphoma by regulating AMPK/mTOR signalling through N4-acetylcytidine modification of SLC30A9. Clin Transl Med 2024; 14:e1747. [PMID: 38961519 PMCID: PMC11222071 DOI: 10.1002/ctm2.1747] [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: 12/16/2023] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Accumulating studies suggested that posttranscriptional modifications exert a vital role in the tumorigenesis of diffuse large B-cell lymphoma (DLBCL). N4-acetylcytidine (ac4C) modification, catalyzed by the N-acetyltransferase 10 (NAT10), was a novel type of chemical modification that improves translation efficiency and mRNA stability. METHODS GEO databases and clinical samples were used to explore the expression and clinical value of NAT10 in DLBCL. CRISPER/Cas9-mediated knockout of NAT10 was performed to determine the biological functions of NAT10 in DLBCL. RNA sequencing, acetylated RNA immunoprecipitation sequencing (acRIP-seq), LC-MS/MS, RNA immunoprecipitation (RIP)-qPCR and RNA stability assays were performed to explore the mechanism by which NAT10 contributed to DLBCL progression. RESULTS Here, we demonstrated that NAT10-mediated ac4C modification regulated the occurrence and progression of DLBCL. Dysregulated N-acetyltransferases expression was found in DLBCL samples. High expression of NAT10 was associated with poor prognosis of DLBCL patients. Deletion of NAT10 expression inhibited cell proliferation and induced G0/G1 phase arrest. Furthermore, knockout of NAT10 increased the sensitivity of DLBCL cells to ibrutinib. AcRIP-seq identified solute carrier family 30 member 9 (SLC30A9) as a downstream target of NAT10 in DLBCL. NAT10 regulated the mRNA stability of SLC30A9 in an ac4C-dependent manner. Genetic silencing of SLC30A9 suppressed DLBCL cell growth via regulating the activation of AMP-activated protein kinase (AMPK) pathway. CONCLUSION Collectively, these findings highlighted the essential role of ac4C RNA modification mediated by NAT10 in DLBCL, and provided insights into novel epigenetic-based therapeutic strategies.
Collapse
Affiliation(s)
- Mengfei Ding
- Department of Hematology, Shandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Zhuoya Yu
- Department of Hematology, Shandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Tiange Lu
- Department of Hematology, Shandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Shunfeng Hu
- Department of Hematology, Shandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Xiangxiang Zhou
- Department of HematologyShandong Provincial Hospital, Affiliated to Shandong First Medical UniversityJinanShandongChina
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xin Wang
- Department of Hematology, Shandong Provincial HospitalShandong UniversityJinanShandongChina
- Department of HematologyShandong Provincial Hospital, Affiliated to Shandong First Medical UniversityJinanShandongChina
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouChina
- Taishan Scholars Program of Shandong ProvinceJinanShandongChina
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongChina
| |
Collapse
|
48
|
Guo Q, Yu W, Tan J, Zhang J, Chen J, Rao S, Guo X, Cai K. Remodelin delays non-small cell lung cancer progression by inhibiting NAT10 via the EMT pathway. Cancer Med 2024; 13:e7283. [PMID: 38826095 PMCID: PMC11145023 DOI: 10.1002/cam4.7283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Lung cancer remains the foremost reason of cancer-related mortality, with invasion and metastasis profoundly influencing patient prognosis. N-acetyltransferase 10 (NAT10) catalyzes the exclusive N (4)-acetylcytidine (ac4C) modification in eukaryotic RNA. NAT10 dysregulation is linked to various diseases, yet its role in non-small cell lung cancer (NSCLC) invasion and metastasis remains unclear. Our study delves into the clinical significance and functional aspects of NAT10 in NSCLC. METHODS We investigated NAT10's clinical relevance using The Cancer Genome Atlas (TCGA) and a group of 98 NSCLC patients. Employing WB, qRT-PCR, and IHC analyses, we assessed NAT10 expression in NSCLC tissues, bronchial epithelial cells (BECs), NSCLC cell lines, and mouse xenografts. Further, knockdown and overexpression techniques (siRNA, shRNA, and plasmid) were employed to evaluate NAT10's effects. A series of assays were carried out, including CCK-8, colony formation, wound healing, and transwell assays, to elucidate NAT10's role in proliferation, invasion, and metastasis. Additionally, we utilized lung cancer patient-derived 3D organoids, mouse xenograft models, and Remodelin (NAT10 inhibitor) to corroborate these findings. RESULTS Our investigations revealed high NAT10 expression in NSCLC tissues, cell lines and mouse xenograft models. High NAT10 level correlated with advanced T stage, lymph node metastasis and poor overall survive. NAT10 knockdown curtailed proliferation, invasion, and migration, whereas NAT10 overexpression yielded contrary effects. Furthermore, diminished NAT10 levels correlated with increased E-cadherin level whereas decreased N-cadherin and vimentin expressions, while heightened NAT10 expression displayed contrasting results. Notably, Remodelin efficiently attenuated NSCLC proliferation, invasion, and migration by inhibiting NAT10 through the epithelial-mesenchymal transition (EMT) pathway. CONCLUSIONS Our data underscore NAT10 as a potential therapeutic target for NSCLC, presenting avenues for targeted intervention against lung cancer through NAT10 inhibition.
Collapse
Affiliation(s)
- Quanwei Guo
- The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Department of Thoracic Surgery, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Weijun Yu
- Bao'an District Hospital for Chronic Diseases Prevention and CureShenzhenChina
| | - Jianfeng Tan
- Department of Thoracic Surgery, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Jianhua Zhang
- Department of Thoracic Surgery, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Jin Chen
- Science and Education Department, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Shuan Rao
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xia Guo
- Center for Clinical Research and Innovation, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
- Shenzhen Key Laboratory of Viral OncologyShenzhenChina
| | - Kaican Cai
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
49
|
Qu Z, Pang X, Mei Z, Li Y, Zhang Y, Huang C, Liu K, Yu S, Wang C, Sun Z, Liu Y, Li X, Jia Y, Dong Y, Lu M, Ju T, Wu F, Huang M, Li N, Dou S, Jiang J, Dong X, Zhang Y, Li W, Yang B, Du W. The positive feedback loop of the NAT10/Mybbp1a/p53 axis promotes cardiomyocyte ferroptosis to exacerbate cardiac I/R injury. Redox Biol 2024; 72:103145. [PMID: 38583415 PMCID: PMC11002668 DOI: 10.1016/j.redox.2024.103145] [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: 02/15/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/09/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of regulated cell death that has been reported to play a central role in cardiac ischemia‒reperfusion (I/R) injury. N-acetyltransferase 10 (NAT10) contributes to cardiomyocyte apoptosis by functioning as an RNA ac4c acetyltransferase, but its role in cardiomyocyte ferroptosis during I/R injury has not been determined. This study aimed to elucidate the role of NAT10 in cardiac ferroptosis as well as the underlying mechanism. The mRNA and protein levels of NAT10 were increased in mouse hearts after I/R and in cardiomyocytes that were exposed to hypoxia/reoxygenation. P53 acted as an endogenous activator of NAT10 during I/R in a transcription-dependent manner. Cardiac overexpression of NAT10 caused cardiomyocyte ferroptosis to exacerbate I/R injury, while cardiomyocyte-specific knockout of NAT10 or pharmacological inhibition of NAT10 with Remodelin had the opposite effects. The inhibition of cardiomyocyte ferroptosis by Fer-1 exerted superior cardioprotective effects against the NAT10-induced exacerbation of post-I/R cardiac damage than the inhibition of apoptosis by emricasan. Mechanistically, NAT10 induced the ac4C modification of Mybbp1a, increasing its stability, which in turn activated p53 and subsequently repressed the transcription of the anti-ferroptotic gene SLC7A11. Moreover, knockdown of Mybbp1a partially abolished the detrimental effects of NAT10 overexpression on cardiomyocyte ferroptosis and cardiac I/R injury. Collectively, our study revealed that p53 and NAT10 interdependently cooperate to form a positive feedback loop that promotes cardiomyocyte ferroptosis to exacerbate cardiac I/R injury, suggesting that targeting the NAT10/Mybbp1a/p53 axis may be a novel approach for treating cardiac I/R.
Collapse
Affiliation(s)
- Zhezhe Qu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiaochen Pang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhongting Mei
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ying Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yaozhi Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chuanhao Huang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Kuiwu Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Shuting Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Changhao Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhiyong Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yingqi Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xin Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yingqiong Jia
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuechao Dong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Meixi Lu
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Tiantian Ju
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Fan Wu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Min Huang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Na Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Shunkang Dou
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jianhao Jiang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xianhui Dong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yi Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wanhong Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Baofeng Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, Harbin, China.
| | - Weijie Du
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, Harbin, China; Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
50
|
Sikder K, Phillips E, Zhong Z, Wang N, Saunders J, Mothy D, Kossenkov A, Schneider T, Nichtova Z, Csordas G, Margulies KB, Choi JC. Perinuclear damage from nuclear envelope deterioration elicits stress responses that contribute to LMNA cardiomyopathy. SCIENCE ADVANCES 2024; 10:eadh0798. [PMID: 38718107 PMCID: PMC11078192 DOI: 10.1126/sciadv.adh0798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/03/2024] [Indexed: 05/12/2024]
Abstract
Mutations in the LMNA gene encoding lamins A/C cause an array of tissue-selective diseases, with the heart being the most commonly affected organ. Despite progress in understanding the perturbations emanating from LMNA mutations, an integrative understanding of the pathogenesis underlying cardiac dysfunction remains elusive. Using a novel conditional deletion model capable of translatome profiling, we observed that cardiomyocyte-specific Lmna deletion in adult mice led to rapid cardiomyopathy with pathological remodeling. Before cardiac dysfunction, Lmna-deleted cardiomyocytes displayed nuclear abnormalities, Golgi dilation/fragmentation, and CREB3-mediated stress activation. Translatome profiling identified MED25 activation, a transcriptional cofactor that regulates Golgi stress. Autophagy is disrupted in the hearts of these mice, which can be recapitulated by disrupting the Golgi. Systemic administration of modulators of autophagy or ER stress significantly delayed cardiac dysfunction and prolonged survival. These studies support a hypothesis wherein stress responses emanating from the perinuclear space contribute to the LMNA cardiomyopathy development.
Collapse
Affiliation(s)
- Kunal Sikder
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Elizabeth Phillips
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Zhijiu Zhong
- Translational Research and Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nadan Wang
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Jasmine Saunders
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - David Mothy
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Andrew Kossenkov
- Bioinformatics Facility, The Wistar Institute Cancer Center, Philadelphia, PA, USA
| | - Timothy Schneider
- Mitocare, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zuzana Nichtova
- Mitocare, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gyorgy Csordas
- Mitocare, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kenneth B. Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason C. Choi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| |
Collapse
|