1
|
Bártová E, Stixová L, Svobodová Kovaříková A. N4-acetylcytidine and other RNA modifications in epitranscriptome: insight into DNA repair and cancer development. Epigenomics 2025; 17:411-422. [PMID: 40040517 PMCID: PMC11980489 DOI: 10.1080/17501911.2025.2473308] [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: 10/03/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
N4-acetylcytidine (ac4C) is a post-transcriptional RNA modification that plays a crucial role in the epitranscriptome, influencing gene expression and cellular function. This modification occurs at the cytosine base, where an acetyl group is installed to the nitrogen at the 4th position (N4). This co-transcription modification affects RNA stability, RNA structure, and translation efficiency. Recent studies have uncovered a potential link between RNA modifications and DNA repair mechanisms, suggesting that ac4C-modified or methylated RNAs may interact with factors involved in DNA repair pathways; thus, influencing the cellular response to DNA damage. Dysregulation of modified RNAs, including ac4C RNA, has been implicated in cancer development, where aberrant levels of these RNAs may contribute to oncogenic transformation by altering genome stability and the expression of key genes regulating cell proliferation, cell cycle progression, and apoptosis. Understanding the dynamics of modified RNAs offers promising insights into the role of epitranscriptome in DNA repair processes and cancer treatment.
Collapse
Affiliation(s)
- Eva Bártová
- Department of Cell Biology and Epigenetics, Institute of Biophysics, the Czech Academy of Sciences, Brno, the Czech Republic
| | - Lenka Stixová
- Department of Cell Biology and Epigenetics, Institute of Biophysics, the Czech Academy of Sciences, Brno, the Czech Republic
| | - Alena Svobodová Kovaříková
- Department of Cell Biology and Epigenetics, Institute of Biophysics, the Czech Academy of Sciences, Brno, the Czech Republic
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Zhao X, Miao J. Biological functions and molecular mechanisms of MORC2 in human diseases. Mol Cells 2025; 48:100166. [PMID: 39637946 PMCID: PMC11731582 DOI: 10.1016/j.mocell.2024.100166] [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: 08/12/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024] Open
Abstract
Microrchidia family CW-type zinc finger 2 (MORC2) is a nuclear protein that has been highly conserved throughout evolution. MORC2 consists of an ATPase domain at the N-terminus, a CW-type zinc finger domain in the middle, and coiled-coil domains at the C-terminus. MORC2 is involved in various important biological processes such as transcriptional regulation, chromatin remodeling, DNA damage repair, and metabolism. Recent studies suggest that MORC2 may serve as a potential biomarker and therapeutic target for hereditary neurological diseases and cancers. However, the exact molecular functions and pathogenic mechanisms of MORC2 in human diseases remain to be explored. In this review, we provide an overview of recent advancements in understanding the molecular functions of MORC2, as well as the characteristics and mechanisms of MORC2-related diseases, which will be valuable for future studies.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Jinfeng Miao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
5
|
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
|
6
|
Xue Z, Xie H, Shan Y, Zhang L, Cheng L, Chen W, Zhu R, Zhang K, Ni H, Zhang Z, You Y, You B. NAT10 inhibition promotes ac4C-dependent ferroptosis to counteract sorafenib resistance in nasopharyngeal carcinoma. Cancer Sci 2024; 115:3256-3272. [PMID: 39038928 PMCID: PMC11447888 DOI: 10.1111/cas.16249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/25/2024] [Accepted: 05/26/2024] [Indexed: 07/24/2024] Open
Abstract
Sorafenib, an anticancer drug, has been shown to induce ferroptosis in cancer cells. However, resistance to sorafenib greatly limits its therapeutic efficacy, and the exact mechanism of resistance is not fully understood. This study investigated the role of N-Acetyltransferase 10 (NAT10) in influencing the anticancer activity of sorafenib in nasopharyngeal carcinoma (NPC) and its molecular mechanism. NAT10 expression was significantly upregulated in NPC. Mechanistically, NAT10 promotes proteins of solute carrier family 7 member 11 (SLC7A11) expression through ac4C acetylation, inhibiting sorafenib-induced ferroptosis in NPC cells. The combined application of sorafenib and the NAT10 inhibitor remodelin significantly inhibits SLC7A11 expression and promotes ferroptosis in NPC cells. In vivo knockout of NAT10 inhibited the growth of sorafenib-resistant NPC. Our findings suggest that NAT10 inhibition might be a promising therapeutic approach to enhance the anticancer activity of sorafenib.
Collapse
Affiliation(s)
- Ziyi Xue
- Nantong UniversityNantongJiangsu ProvinceChina
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Haijing Xie
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
- Department of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Ying Shan
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
- Department of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Lin Zhang
- Haimen People's HospitalNantongJiangsu ProvinceChina
| | - Lin Cheng
- Nantong UniversityNantongJiangsu ProvinceChina
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Wenyue Chen
- Nantong UniversityNantongJiangsu ProvinceChina
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Rui Zhu
- Nantong UniversityNantongJiangsu ProvinceChina
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Kaiwen Zhang
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
- Department of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Haosheng Ni
- Department of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Zhenxin Zhang
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
- Department of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Yiwen You
- Nantong UniversityNantongJiangsu ProvinceChina
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| | - Bo You
- Nantong UniversityNantongJiangsu ProvinceChina
- Institute of Otolaryngology Head and Neck SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsu ProvinceChina
| |
Collapse
|
7
|
Engbrecht M, Grundei D, Dilger A, Wiedemann H, Aust AK, Baumgärtner S, Helfrich S, Kergl-Räpple F, Bürkle A, Mangerich A. Monitoring nucleolar-nucleoplasmic protein shuttling in living cells by high-content microscopy and automated image analysis. Nucleic Acids Res 2024; 52:e72. [PMID: 39036969 PMCID: PMC11347172 DOI: 10.1093/nar/gkae598] [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: 09/25/2023] [Revised: 05/25/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024] Open
Abstract
The nucleolus has core functions in ribosome biosynthesis, but also acts as a regulatory hub in a plethora of non-canonical processes, including cellular stress. Upon DNA damage, several DNA repair factors shuttle between the nucleolus and the nucleoplasm. Yet, the molecular mechanisms underlying such spatio-temporal protein dynamics remain to be deciphered. Here, we present a novel imaging platform to investigate nucleolar-nucleoplasmic protein shuttling in living cells. For image acquisition, we used a commercially available automated fluorescence microscope and for image analysis, we developed a KNIME workflow with implementation of machine learning-based tools. We validated the method with different nucleolar proteins, i.e., PARP1, TARG1 and APE1, by monitoring their shuttling dynamics upon oxidative stress. As a paradigm, we analyzed PARP1 shuttling upon H2O2 treatment in combination with a range of pharmacological inhibitors in a novel reporter cell line. These experiments revealed that inhibition of SIRT7 results in a loss of nucleolar PARP1 localization. Finally, we unraveled specific differences in PARP1 shuttling dynamics after co-treatment with H2O2 and different clinical PARP inhibitors. Collectively, this work delineates a highly sensitive and versatile bioimaging platform to investigate swift nucleolar-nucleoplasmic protein shuttling in living cells, which can be employed for pharmacological screening and in-depth mechanistic analyses.
Collapse
Affiliation(s)
- Marina Engbrecht
- Molecular Toxicology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - David Grundei
- Molecular Toxicology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Asisa M Dilger
- Nutritional Toxicology, Institute of Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
| | - Hannah Wiedemann
- Molecular Toxicology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Ann-Kristin Aust
- Molecular Toxicology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Sarah Baumgärtner
- Molecular Toxicology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | | | | | - Alexander Bürkle
- Molecular Toxicology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Aswin Mangerich
- Molecular Toxicology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
- Nutritional Toxicology, Institute of Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
| |
Collapse
|
8
|
Mohapatra B, Pakala SB. Emerging roles of the chromatin remodeler MORC2 in cancer metabolism. Med Oncol 2024; 41:221. [PMID: 39117768 DOI: 10.1007/s12032-024-02464-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
Cancer is characterized by metabolic reprogramming in cancer cells, which is crucial for tumorigenesis. The highly deregulated chromatin remodeler MORC2 contributes to cell proliferation, invasion, migration, DNA repair, and chemoresistance. MORC2 also plays a key role in metabolic reprogramming, including lipogenesis, glucose, and glutamine metabolism. A recent study showed that MORC2-regulated glucose metabolism affects the expression of E-cadherin, a crucial protein in the epithelial-to-mesenchymal transition. This review discusses recent developments in MORC2 regulated cancer cell metabolism and its role in cancer progression.
Collapse
Affiliation(s)
- Bibhukalyan Mohapatra
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500 046, India
| | - Suresh B Pakala
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500 046, India.
| |
Collapse
|
9
|
Achour C, Oberdoerffer S. NAT10 and cytidine acetylation in mRNA: intersecting paths in development and disease. Curr Opin Genet Dev 2024; 87:102207. [PMID: 38820741 DOI: 10.1016/j.gde.2024.102207] [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: 01/12/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 06/02/2024]
Abstract
N4-acetylcytidine (ac4C) is an RNA modification that is catalyzed by the enzyme NAT10. Constitutively found in tRNA and rRNA, ac4C displays a dynamic presence in mRNA that is shaped by developmental and induced shifts in NAT10 levels. However, deciphering ac4C functions in mRNA has been hampered by its context-dependent influences in translation and the complexity of isolating effects on specific mRNAs from other NAT10 activities. Recent advances have begun to overcome these obstacles by leveraging natural variations in mRNA acetylation in cancer, developmental transitions, and immune responses. Here, we synthesize the current literature with a focus on nuances that may fuel the perception of cellular discrepancies toward the development of a cohesive model of ac4C function in mRNA.
Collapse
Affiliation(s)
- Cyrinne Achour
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Shalini Oberdoerffer
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA.
| |
Collapse
|
10
|
Dalhat MH, Narayan S, Serio H, Arango D. Dissecting the oncogenic properties of essential RNA-modifying enzymes: a focus on NAT10. Oncogene 2024; 43:1077-1086. [PMID: 38409550 PMCID: PMC11092965 DOI: 10.1038/s41388-024-02975-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/28/2024]
Abstract
Chemical modifications of ribonucleotides significantly alter the physicochemical properties and functions of RNA. Initially perceived as static and essential marks in ribosomal RNA (rRNA) and transfer RNA (tRNA), recent discoveries unveiled a dynamic landscape of RNA modifications in messenger RNA (mRNA) and other regulatory RNAs. These findings spurred extensive efforts to map the distribution and function of RNA modifications, aiming to elucidate their distribution and functional significance in normal cellular homeostasis and pathological states. Significant dysregulation of RNA modifications is extensively documented in cancers, accentuating the potential of RNA-modifying enzymes as therapeutic targets. However, the essential role of several RNA-modifying enzymes in normal physiological functions raises concerns about potential side effects. A notable example is N-acetyltransferase 10 (NAT10), which is responsible for acetylating cytidines in RNA. While emerging evidence positions NAT10 as an oncogenic factor and a potential target in various cancer types, its essential role in normal cellular processes complicates the development of targeted therapies. This review aims to comprehensively analyze the essential and oncogenic properties of NAT10. We discuss its crucial role in normal cell biology and aging alongside its contribution to cancer development and progression. We advocate for agnostic approaches to disentangling the intertwined essential and oncogenic functions of RNA-modifying enzymes. Such approaches are crucial for understanding the full spectrum of RNA-modifying enzymes and imperative for designing effective and safe therapeutic strategies.
Collapse
Affiliation(s)
- Mahmood H Dalhat
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Sharath Narayan
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, USA
| | - Hannah Serio
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Daniel Arango
- Department of Pharmacology, Northwestern University, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
11
|
Xi M, Yan Y, Duan S, Li T, Szeto IMY, Zhao A. Short-chain fatty acids in breast milk and their relationship with the infant gut microbiota. Front Microbiol 2024; 15:1356462. [PMID: 38440144 PMCID: PMC10909814 DOI: 10.3389/fmicb.2024.1356462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
Introduction The short-chain fatty acids (SCFAs) contained in breast milk play a key role in infant growth, affecting metabolism and enhancing intestinal immunity by regulating inflammation. Methods In order to examine the associations between the microbiota and SCFA levels in breast milk, and explore the roles of SCFAs in regulating the infant gut microbiota, we enrolled 50 paired mothers and infants and collected both breast milk and infant fecal samples. Breast milk SCFA contents were determined by UPLC-MS, and whole genome shotgun sequencing was applied to determine the microbial composition of breast milk and infant feces. The SCFA levels in breast milk were grouped into tertiles as high, medium, or low, and the differences of intestinal microbiota and KEGG pathways were compared among groups. Results The results demonstrated that breast milk butyric acid (C4) is significantly associated with Clostridium leptum richness in breastmilk. Additionally, the specific Bifidobacterium may have an interactive symbiosis with the main species of C4-producing bacteria in human milk. Women with a low breast milk C4 tertile are associated with a high abundance of Salmonella and Salmonella enterica in their infants' feces. KEGG pathway analysis further showed that the content of C4 in breast milk is significantly correlated with the infants' metabolic pathways of lysine and arginine biosynthesis. Discussion This study suggests that interactive symbiosis of the microbiota exists in breast milk. Certain breast milk microbes could be beneficial by producing C4 and further influence the abundance of certain gut microbes in infants, playing an important role in early immune and metabolic development.
Collapse
Affiliation(s)
- Menglu Xi
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Yalu Yan
- Inner Mongolia Yili Industrial Group Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot, China
| | - Sufang Duan
- Inner Mongolia Yili Industrial Group Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot, China
| | - Ting Li
- Inner Mongolia Yili Industrial Group Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot, China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Ai Zhao
- Vanke School of Public Health, Tsinghua University, Beijing, China
| |
Collapse
|
12
|
Kołacz K, Robaszkiewicz A. PARP1 at the crossroad of cellular senescence and nucleolar processes. Ageing Res Rev 2024; 94:102206. [PMID: 38278370 DOI: 10.1016/j.arr.2024.102206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/09/2024] [Accepted: 01/22/2024] [Indexed: 01/28/2024]
Abstract
Senescent cells that occur in response to telomere shortening, oncogenes, extracellular and intracellular stress factors are characterized by permanent cell cycle arrest, the morphological and structural changes of the cell that include the senescence-associated secretory phenotype (SASP) and nucleoli rearrangement. The associated DNA lesions induce DNA damage response (DDR), which activates the DNA repair protein - poly-ADP-ribose polymerase 1 (PARP1). This protein consumes NAD+ to synthesize ADP-ribose polymer (PAR) on its own protein chain and on other interacting proteins. The involvement of PARP1 in nucleoli processes, such as rRNA transcription and ribosome biogenesis, the maintenance of heterochromatin and nucleoli structure, as well as controlling the crucial DDR protein release from the nucleoli to nucleus, links PARP1 with cellular senescence and nucleoli functioning. In this review we describe and discuss the impact of PARP1-mediated ADP-ribosylation on early cell commitment to senescence with the possible role of senescence-induced PARP1 transcriptional repression and protein degradation on nucleoli structure and function. The cause-effect interplay between PARP1 activation/decline and nucleoli functioning during senescence needs to be studied in detail.
Collapse
Affiliation(s)
- Kinga Kołacz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, Banacha 12 /16, 90-237 Lodz, Poland.
| | - Agnieszka Robaszkiewicz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research (IFBR), 600 5th Street South, St. Petersburgh, FL 33701, USA.
| |
Collapse
|
13
|
Zhang Y, Lei Y, Dong Y, Chen S, Sun S, Zhou F, Zhao Z, Chen B, Wei L, Chen J, Meng Z. Emerging roles of RNA ac4C modification and NAT10 in mammalian development and human diseases. Pharmacol Ther 2024; 253:108576. [PMID: 38065232 DOI: 10.1016/j.pharmthera.2023.108576] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 01/13/2024]
Abstract
RNA ac4C modification is a novel and rare chemical modification observed in mRNA. Traditional biochemical studies had primarily associated ac4C modification with tRNA and rRNA until in 2018, Arango D et al. first reported the presence of ac4C modification on mRNA and demonstrated its critical role in mRNA stability and translation regulation. Furthermore, they established that the ac4C modification on mRNA is mediated by the classical N-acetyltransferase NAT10. Subsequent studies have underscored the essential implications of NAT10 and mRNA ac4C modification across both physiological and pathological regulatory processes. In this review, we aimed to explore the discovery history of RNA ac4C modification, its detection methods, and its regulatory mechanisms in disease and physiological development. We offer a forward-looking examination and discourse concerning the employment of RNA ac4C modification as a prospective therapeutic strategy across diverse diseases. Furthermore, we comprehensively summarize the functions and mechanisms of NAT10 in gene expression regulation and pathogenesis independent of RNA ac4C modification.
Collapse
Affiliation(s)
- Yigan Zhang
- Institute of Biomedical Research, Department of Infectious Diseases, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei rovincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Yumei Lei
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yanbin Dong
- Institute of Biophysics, Chinese Academy of Sciences, Key Laboratory of Nucleic Acid Biology, Chinese Academy of Sciences, Beijing, China
| | - Shuwen Chen
- School of Biomedical Engineering, Hubei University of Medicine, Shiyan, China
| | - Siyuan Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Fange Zhou
- The First Clinical School of Hubei University of Medicine, Shiyan, China
| | - Zhiwen Zhao
- Department of Emergency Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Lv Wei
- Institute of Biophysics, Chinese Academy of Sciences, Key Laboratory of Nucleic Acid Biology, Chinese Academy of Sciences, Beijing, China.
| | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Zhongji Meng
- Institute of Biomedical Research, Department of Infectious Diseases, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei rovincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| |
Collapse
|
14
|
Chutani N, Ragula S, Syed K, Pakala SB. Novel Insights into the Role of Chromatin Remodeler MORC2 in Cancer. Biomolecules 2023; 13:1527. [PMID: 37892209 PMCID: PMC10605154 DOI: 10.3390/biom13101527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
A newly discovered chromatin remodeler, MORC2, is a Microrchidia (MORC) family member. MORC2 acts as a chromatin remodeler by binding to the DNA and changing chromatin conformation using its ATPase domain. MORC2 is highly expressed in a variety of human cancers. It controls diverse signaling pathways essential for cancer development through its target genes and interacting partners. MORC2 promotes cancer cells' growth, invasion, and migration by regulating the expression of genes involved in these processes. MORC2 is localized primarily in the nucleus and is also found in the cytoplasm. In the cytoplasm, MORC2 interacts with adenosine triphosphate (ATP)-citrate lyase (ACLY) to promote lipogenesis and cholesterogenesis in cancer. In the nucleus, MORC2 interacts with the transcription factor c-Myc to control the transcription of genes involved in glucose metabolism to drive cancer cell migration and invasion. Furthermore, MORC2 recruits on to the promoters of tumor suppressor genes to repress their transcription and expression to promote oncogenesis. In addition to its crucial function in oncogenesis, it plays a vital role in DNA repair. Overall, this review concisely summarizes the current knowledge about MORC2-regulated molecular pathways involved in cancer.
Collapse
Affiliation(s)
- Namita Chutani
- Biology Division, Indian Institute of Science Education and Research (IISER) Tirupati, Mangalam, Tirupati 517 507, India;
| | - Sandhya Ragula
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500 046, India;
| | - Khajamohiddin Syed
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture and Engineering, University of Zululand, KwaDlangezwa 3886, South Africa;
| | - Suresh B. Pakala
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500 046, India;
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture and Engineering, University of Zululand, KwaDlangezwa 3886, South Africa;
| |
Collapse
|
15
|
Li Z, Luo A, Xie B. The Complex Network of ADP-Ribosylation and DNA Repair: Emerging Insights and Implications for Cancer Therapy. Int J Mol Sci 2023; 24:15028. [PMID: 37834477 PMCID: PMC10573881 DOI: 10.3390/ijms241915028] [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: 08/21/2023] [Revised: 09/23/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
ADP-ribosylation is a post-translational modification of proteins that plays a key role in various cellular processes, including DNA repair. Recently, significant progress has been made in understanding the mechanism and function of ADP-ribosylation in DNA repair. ADP-ribosylation can regulate the recruitment and activity of DNA repair proteins by facilitating protein-protein interactions and regulating protein conformations. Moreover, ADP-ribosylation can influence additional post-translational modifications (PTMs) of proteins involved in DNA repair, such as ubiquitination, methylation, acetylation, phosphorylation, and SUMOylation. The interaction between ADP-ribosylation and these additional PTMs can fine-tune the activity of DNA repair proteins and ensure the proper execution of the DNA repair process. In addition, PARP inhibitors have been developed as a promising cancer therapeutic strategy by exploiting the dependence of certain cancer types on the PARP-mediated DNA repair pathway. In this paper, we review the progress of ADP-ribosylation in DNA repair, discuss the crosstalk of ADP-ribosylation with additional PTMs in DNA repair, and summarize the progress of PARP inhibitors in cancer therapy.
Collapse
Affiliation(s)
| | - Aiqin Luo
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Bingteng Xie
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
16
|
Desingu PA, Mishra S, Dindi L, Srinivasan S, Rajmani RS, Ravi V, Tamta AK, Raghu S, Murugasamy K, Pandit AS, Sundaresan NR. PARP1 inhibition protects mice against Japanese encephalitis virus infection. Cell Rep 2023; 42:113103. [PMID: 37676769 DOI: 10.1016/j.celrep.2023.113103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 05/20/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023] Open
Abstract
Japanese encephalitis (JE) is a vector-borne viral disease that causes acute encephalitis in children. Although vaccines have been developed against the JE virus (JEV), no effective antiviral therapy exists. Our study shows that inhibition of poly(ADP-ribose) polymerase 1 (PARP1), an NAD+-dependent (poly-ADP) ribosyl transferase, protects against JEV infection. Interestingly, PARP1 is critical for JEV pathogenesis in Neuro-2a cells and mice. Small molecular inhibitors of PARP1, olaparib, and 3-aminobenzamide (3-AB) significantly reduce clinical signs and viral load in the serum and brains of mice and improve survival. PARP1 inhibition confers protection against JEV infection by inhibiting autophagy. Mechanistically, upon JEV infection, PARP1 PARylates AKT and negatively affects its phosphorylation. In addition, PARP1 transcriptionally upregulates PTEN, the PIP3 phosphatase, negatively regulating AKT. PARP1-mediated AKT inactivation promotes autophagy and JEV pathogenesis by increasing the FoxO activity. Thus, our findings demonstrate PARP1 as a potential mediator of JEV pathogenesis that can be effectively targeted for treating JE.
Collapse
Affiliation(s)
- Perumal Arumugam Desingu
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India.
| | - Sneha Mishra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India
| | - Lavanya Dindi
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India
| | - Shalini Srinivasan
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India
| | - Raju S Rajmani
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India
| | - Venkatraman Ravi
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India
| | - Ankit Kumar Tamta
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India
| | - Sukanya Raghu
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India
| | - Krishnega Murugasamy
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India
| | - Anwit Shriniwas Pandit
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India
| | - Nagalingam R Sundaresan
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, India.
| |
Collapse
|
17
|
Luo J, Cao J, Chen C, Xie H. Emerging role of RNA acetylation modification ac4C in diseases: Current advances and future challenges. Biochem Pharmacol 2023; 213:115628. [PMID: 37247745 DOI: 10.1016/j.bcp.2023.115628] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 05/31/2023]
Abstract
The oldest known highly conserved modification of RNA, N4-acetylcytidine, is widely distributed from archaea to eukaryotes and acts as a posttranscriptional chemical modification of RNA, contributing to the correct reading of specific nucleotide sequences during translation, stabilising mRNA and improving transcription efficiency. Yeast Kre33 and human NAT10, the only known authors of ac4C, modify tRNA with the help of the Tan1/THUMPD1 adapter to stabilise its structure. Currently, the mRNA for N4-acetylcytidine (ac4C), catalysed by NAT10 (N-acetyltransferase 10), has been implicated in a variety of human diseases, particularly cancer. This article reviews advances in the study of ac4C modification of RNA and the ac4C-related gene NAT10 in normal physiological cell development, cancer, premature disease and viral infection and discusses its therapeutic promise and future research challenges.
Collapse
Affiliation(s)
- Jie Luo
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jingsong Cao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Institute of Clinical Medicine, University of South China, Hengyang 421001, China
| | - Cong Chen
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Haitao Xie
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
18
|
Long Y, Ren Y, Wei Q, Mobet Y, Liu Y, Zhao H, Liu T, Cheng L, Yi P. NAT10-mediated RNA acetylation enhances HNRNPUL1 mRNA stability to contribute cervical cancer progression. Int J Med Sci 2023; 20:1079-1090. [PMID: 37484809 PMCID: PMC10357443 DOI: 10.7150/ijms.83828] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/06/2023] [Indexed: 07/25/2023] Open
Abstract
N4-acetylcytidine (ac4C) is a lately discovered nucleotide modification that has been shown to be closely implicated in cancer. N-acetyltransferase10(NAT10) acts as an enzyme that regulates mRNA acetylation modifications. Currently, the role of NAT10-mediated RNA acetylation modification in cervical cancer remains to be elucidated. On the basis of transcriptome analysis of TCGA and GEO open datasets (GSE52904, GSE29570, GSE122697), NAT10 is upregulated in cervical cancer tissues and correlated with poor prognosis. Knockdown of NAT10 suppressed the cell proliferation, invasion, and migration of cervical cancer cells. The in vivo oncogenic function of NAT10 was also confirmed in xenograft models. Combined RNA-seq and acRIP-seq analysis revealed HNRNPUL1 as the target of NAT10 in cervical cancer. NAT10 positively regulate HNRNPUL1 expression by promoting ac4C modification and stability of HNRNPUL1 mRNA. Furthermore, depletion of HNRNPUL1 suppressed the cell division, invasion, and migration of cervical cancer. HNRNPUL1 overexpression partially restored cellular function in cervical cancer cells with NAT10 knockdown. Thus, this study demonstrates that NAT10 contributes to cervical cancer progression by enhancing HNRNPUL1 mRNA stability via ac4C modification, and NAT10-ac4C-HNRNPUL1 axis might be a potential target for cervical cancer therapy.
Collapse
Affiliation(s)
- Yingfei Long
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yifei Ren
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Qinglv Wei
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Youchaou Mobet
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yujiao Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Hongyan Zhao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Tao Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Lei Cheng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| |
Collapse
|
19
|
Dalhat MH, Choudhry H, Khan MI. NAT10, an RNA Cytidine Acetyltransferase, Regulates Ferroptosis in Cancer Cells. Antioxidants (Basel) 2023; 12:antiox12051116. [PMID: 37237981 DOI: 10.3390/antiox12051116] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Recently, we reported that N-acetyltransferase 10 (NAT10) regulates fatty acid metabolism through ac4C-dependent RNA modification of key genes in cancer cells. During this work, we noticed ferroptosis as one of the most negatively enriched pathways among other pathways in NAT10-depleted cancer cells. In the current work, we explore the possibility of whether NAT10 acts as an epitranscriptomic regulator of the ferroptosis pathway in cancer cells. Global ac4C levels and expression of NAT10 with other ferroptosis-related genes were assessed via dotblot and RT-qPCR, respectively. Flow cytometry and biochemical analysis were used to assess oxidative stress and ferroptosis features. The ac4C-mediated mRNA stability was conducted using RIP-PCR and mRNA stability assay. Metabolites were profiled using LC-MS/MS. Our results showed significant downregulation in expression of essential genes related to ferroptosis, namely SLC7A11, GCLC, MAP1LC3A, and SLC39A8 in NAT10-depleted cancer cells. Further, we noticed a reduction in cystine uptake and reduced GSH levels, along with elevated ROS, and lipid peroxidation levels in NAT10-depleted cells. Consistently, overproduction of oxPLs, as well as increased mitochondrial depolarization and decreased activities of antioxidant enzymes, support the notion of ferroptosis induction in NAT10-depleted cancer cells. Mechanistically, a reduced ac4C level shortens the half-life of GCLC and SLC7A11 mRNA, resulting in low levels of intracellular cystine and reduced GSH, failing to detoxify ROS, and leading to increased cellular oxPLs, which facilitate ferroptosis induction. Collectively, our findings suggest that NAT10 restrains ferroptosis by stabilizing the SLC7A11 mRNA transcripts in order to avoid oxidative stress that induces oxidation of phospholipids to initiate ferroptosis.
Collapse
Affiliation(s)
- Mahmood Hassan Dalhat
- Department of Biochemistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hani Choudhry
- Department of Biochemistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad Imran Khan
- Department of Biochemistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
20
|
MORC2 and MAX contributes to the expression of glycolytic enzymes, breast cancer cell proliferation and migration. Med Oncol 2023; 40:102. [PMID: 36802305 DOI: 10.1007/s12032-023-01974-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/05/2023] [Indexed: 02/23/2023]
Abstract
Cancer cell proliferation is a high energy demanding process, where the cancer cells acquire energy by high rates of glycolysis, and this phenomenon is known as the "Warburg effect". Microrchidia 2 (MORC2), an emerging chromatin remodeler, is over expressed in several cancers including breast cancer and found to promote cancer cell proliferation. However, the role of MORC2 in glucose metabolism in cancer cells remains unexplored. In this study, we report that MORC2 interacts indirectly with the genes involved in glucose metabolism via transcription factors MAX (MYC-associated factor X) and MYC. We also found that MORC2 co-localizes and interacts with MAX. Further, we observed a positive correlation of expression of MORC2 with glycolytic enzymes Hexokinase 1 (HK1), Lactate dehydrogenase A (LDHA) and Phosphofructokinase platelet (PFKP) type in multiple cancers. Surprisingly, the knockdown of either MORC2 or MAX not only decreased the expression of glycolytic enzymes but also inhibited breast cancer cell proliferation and migration. Together, these results demonstrate the involvement of the MORC2/MAX signaling axis in the expression of glycolytic enzymes and breast cancer cell proliferation and migration.
Collapse
|
21
|
Zhu T, Zheng JY, Huang LL, Wang YH, Yao DF, Dai HB. Human PARP1 substrates and regulators of its catalytic activity: An updated overview. Front Pharmacol 2023; 14:1137151. [PMID: 36909172 PMCID: PMC9995695 DOI: 10.3389/fphar.2023.1137151] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) is a key DNA damage sensor that is recruited to damaged sites after DNA strand breaks to initiate DNA repair. This is achieved by catalyzing attachment of ADP-ribose moieties, which are donated from NAD+, on the amino acid residues of itself or other acceptor proteins. PARP inhibitors (PARPi) that inhibit PARP catalytic activity and induce PARP trapping are commonly used for treating BRCA1/2-deficient breast and ovarian cancers through synergistic lethality. Unfortunately, resistance to PARPi frequently occurs. In this review, we present the novel substrates and regulators of the PARP1-catalyzed poly (ADP-ribosyl)ation (PARylatison) that have been identified in the last 3 years. The overall aim is the presentation of protein interactions of potential therapeutic intervention for overcoming the resistance to PARPi.
Collapse
Affiliation(s)
- Tao Zhu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ju-Yan Zheng
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Ling Huang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan-Hong Wang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Di-Fei Yao
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Bin Dai
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|