1
|
Liu W, Ma ZC, Zhang S, Gang FY, Ji TT, Gu YH, Xie NB, Gu SY, Guo X, Feng T, Liu Y, Xiong J, Yuan BF. Direct single-nucleotide resolution sequencing of DNA 5-methylcytosine using engineered DNA methyltransferase-mediated CMD-seq. Chem Sci 2025; 16:8788-8799. [PMID: 40276634 PMCID: PMC12015181 DOI: 10.1039/d5sc01211b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
5-Methylcytosine (5mC) is a crucial epigenetic modification in the mammalian genome, primarily occurring at CG dinucleotides. Accurate localization of 5mC is essential for understanding its functional significance. In this study, we discovered a novel DNA methyltransferase, designated M.MedI, from the bacterium Mycoplasmopsis edwardii. M.MedI exhibits carboxymethylation activity towards cytosines in CG sites in DNA. We further engineered a variant of M.MedI by mutating its critical active site residue 377 asparagine (N) to lysine (K), resulting in M.MedI-N377K. This engineered M.MedI-N377K enzyme demonstrated superior carboxymethylation activity towards cytosines in CG sites in both unmethylated and hemi-methylated DNA. Utilizing the newly identified M.MedI-N377K methyltransferase, we developed a novel method, engineered DNA methyltransferase-mediated carboxymethylation deamination sequencing (CMD-seq), for the stoichiometric detection of 5mC in DNA at single-nucleotide resolution. In CMD-seq, M.MedI-N377K efficiently transfers a carboxymethyl group to cytosines in CG sites in the presence of carboxy-S-adenosyl-l-methionine (caSAM), generating 5-carboxymethylcytosine (5camC). Subsequent treatment with the deaminase A3A deaminates 5mC to form thymine (T), which pairs with adenine (A) and is read as T, while 5camC remains unchanged, pairing with guanine (G) and being read as cytosine (C) during sequencing. We successfully applied CMD-seq to quantify 5mC sites in the promoters of tumor suppressor genes RASSF1A and SHOX2 in human lung cancer tissue and adjacent normal tissue. The quantification results were highly comparable to those obtained using traditional bisulfite sequencing. Overall, CMD-seq provides a valuable tool for bisulfite-free, single-nucleotide resolution, and quantitative detection of 5mC in limited DNA samples.
Collapse
Affiliation(s)
- Wei Liu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
- Research Center of Public Health, Renmin Hospital of Wuhan University, Wuhan University Wuhan 430060 China
| | - Zhao-Cheng Ma
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Shan Zhang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Fang-Yin Gang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Tong-Tong Ji
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Yao-Hua Gu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
- School of Nursing, Wuhan University Wuhan 430071 China
| | - Neng-Bin Xie
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Shu-Yi Gu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Xia Guo
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Tian Feng
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Yu Liu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
- Hubei Key Laboratory of Tumor Biological Behaviors, Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Jun Xiong
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Bi-Feng Yuan
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
- Research Center of Public Health, Renmin Hospital of Wuhan University, Wuhan University Wuhan 430060 China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Wuhan University Wuhan 430072 China
- Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, Wuhan University Wuhan 430071 China
| |
Collapse
|
2
|
Lu B, Guo Z, Liu X, Ni Y, Xu L, Huang J, Li T, Feng T, Li R, Deng X. Comprehensive comparison of the third-generation sequencing tools for bacterial 6mA profiling. Nat Commun 2025; 16:3982. [PMID: 40295502 PMCID: PMC12037826 DOI: 10.1038/s41467-025-59187-2] [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: 06/18/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
DNA N6-methyladenine (6mA) serves as an intrinsic and principal epigenetic marker in prokaryotes, impacting various biological processes. To date, limited advanced sequencing technologies and analyzing tools are available for bacterial DNA 6mA. Here, we evaluate eight tools designed for the 6mA identification or de novo methylation detection. This assessment includes Nanopore (R9 and R10), Single-Molecule Real-Time (SMRT) Sequencing, and cross-reference with 6mA-IP-seq and DR-6mA-seq. Our multi-dimensional evaluation report encompasses motif discovery, site-level accuracy, single-molecule accuracy, and outlier detection across six bacteria strains. While most tools correctly identify motifs, their performance varies at single-base resolution, with SMRT and Dorado consistently delivering strong performance. Our study indicates that existing tools cannot accurately detect low-abundance methylation sites. Additionally, we introduce an optimized method for advancing 6mA prediction, which substantially improves the detection performance of Dorado. Overall, our study provides a robust and detailed examination of computational tools for bacterial 6mA profiling, highlighting insights for further tool enhancement and epigenetic research.
Collapse
Grants
- Shenzhen Science and Technology Fund, JCYJ20210324134000002, recipient: Xin Deng Guangdong Major Project of Basic and Applied Basic Research, 2020B0301030005, recipient: Xin Deng National Natural Science Foundation of China, 32172358, recipient: Xin Deng General Research Funds of Hong Kong, 11103221, recipient: Xin Deng General Research Funds of Hong Kong, 11102223, recipient: Xin Deng General Research Funds of Hong Kong, 11101722, recipient: Xin Deng
Collapse
Affiliation(s)
- Beifang Lu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Zhihao Guo
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong SAR, China
| | - Xudong Liu
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong SAR, China
| | - Ying Ni
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong SAR, China
| | - Letong Xu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Jiadai Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Tianmin Li
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Tongtong Feng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Runsheng Li
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong SAR, China.
- Tung Biomedical Sciences Center, City University of Hong Kong, Hong Kong, China.
| | - Xin Deng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.
- Tung Biomedical Sciences Center, City University of Hong Kong, Hong Kong, China.
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, Guangdong, China.
| |
Collapse
|
3
|
Steadman CR, Small EM, Banerjee S, Gonzalez-Esquer CR, Pacheco S, Twary SN. Best practices for methylome characterization in novel species: a case study in the microalgae Microchloropsis. Commun Biol 2025; 8:648. [PMID: 40269108 PMCID: PMC12019312 DOI: 10.1038/s42003-025-08027-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Microalgae remain an important feedstock in the circular bioeconomy. The discovery of new species combined with advanced biotechnology drives optimization of performance predicated on deep knowledge of algal genomics and phenotype. Understanding the contribution of epigenetic processes to algal function provides insight and better approaches for achieving production goals. Here, we provide a methodological framework for investigating epigenetic modifications in new species, including analysis of state-of-the-art techniques, and best practices for discerning novel modifications, focusing on variants of DNA methylation. Further, we demonstrate that specific forms of DNA methylation are overlooked by traditional epigenetic analysis strategies. Using high-throughput, lower cost techniques, we provide several pieces of evidence demonstrating Microchloropsis gaditana and M. salina (formerly Nannochloropsis), two candidate feedstock species, lack the most ubiquitous forms of eukaryotic DNA methylation (5mC and 5hmC) and instead employ N6-adenine methylation (6mA), commonly found in bacteria, in their genomes. Interestingly, transcriptionally diverse physiological conditions do not elicit differential 6mA methylation, suggesting the presence of 6mA may provide stability and protection of the genome. These collective discoveries illuminate not only an exciting avenue for improving feedstock genetic drift, stability, and culture health for bioproduction but also an ideal model species to study other epigenetic processes in microalgae.
Collapse
Affiliation(s)
- Christina R Steadman
- Genomics & Bioanalytics Group, Los Alamos National Laboratory, Los Alamos, NM, USA.
- Climate, Ecology & Environment Group, Los Alamos National Laboratory, Los Alamos, NM, USA.
| | - Eric M Small
- Genomics & Bioanalytics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
- Climate, Ecology & Environment Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Shounak Banerjee
- Genomics & Bioanalytics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | | | - Sara Pacheco
- Microbial & Biome Science Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Scott N Twary
- Microbial & Biome Science Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| |
Collapse
|
4
|
Walker J, Zhang J, Liu Y, Xu S, Yu Y, Vickers M, Ouyang W, Tálas J, Dolan L, Nakajima K, Feng X. Extensive N4 cytosine methylation is essential for Marchantia sperm function. Cell 2025:S0092-8674(25)00287-9. [PMID: 40209706 DOI: 10.1016/j.cell.2025.03.014] [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: 02/09/2022] [Revised: 06/30/2024] [Accepted: 03/07/2025] [Indexed: 04/12/2025]
Abstract
N4-methylcytosine (4mC) is an important DNA modification in prokaryotes, but its relevance and even its presence in eukaryotes have been mysterious. Here we show that spermatogenesis in the liverwort Marchantia polymorpha involves two waves of extensive DNA methylation reprogramming. First, 5-methylcytosine (5mC) expands from transposons to the entire genome. Notably, the second wave installs 4mC throughout genic regions, covering over 50% of CG sites in sperm. 4mC requires a methyltransferase (MpDN4MT1a) that is specifically expressed during late spermiogenesis. Deletion of MpDN4MT1a alters the sperm transcriptome, causes sperm swimming and fertility defects, and impairs post-fertilization development. Our results reveal extensive 4mC in a eukaryote, identify a family of eukaryotic methyltransferases, and elucidate the biological functions of 4mC in reproductive development, thereby expanding the repertoire of functional eukaryotic DNA modifications.
Collapse
Affiliation(s)
- James Walker
- Department of Cell and Developmental Biology, John Innes Centre, Norwich NR4 7UH, UK
| | - Jingyi Zhang
- Department of Cell and Developmental Biology, John Innes Centre, Norwich NR4 7UH, UK; Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Yalin Liu
- Department of Cell and Developmental Biology, John Innes Centre, Norwich NR4 7UH, UK; Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Shujuan Xu
- Institute of Science and Technology Austria, Klosterneuburg 3400, Austria
| | - Yiming Yu
- Institute of Science and Technology Austria, Klosterneuburg 3400, Austria
| | - Martin Vickers
- Department of Cell and Developmental Biology, John Innes Centre, Norwich NR4 7UH, UK
| | - Weizhi Ouyang
- Institute of Science and Technology Austria, Klosterneuburg 3400, Austria
| | - Judit Tálas
- Department of Cell and Developmental Biology, John Innes Centre, Norwich NR4 7UH, UK
| | - Liam Dolan
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, Vienna 1030, Austria
| | - Keiji Nakajima
- Nara Institute of Science and Technology, Nara 630-0192, Japan
| | - Xiaoqi Feng
- Department of Cell and Developmental Biology, John Innes Centre, Norwich NR4 7UH, UK; Institute of Science and Technology Austria, Klosterneuburg 3400, Austria.
| |
Collapse
|
5
|
Guo X, Wu J, Ji TT, Wang M, Zhang S, Xiong J, Gang FY, Liu W, Gu YH, Liu Y, Xie NB, Yuan BF. Orthologous mammalian A3A-mediated single-nucleotide resolution sequencing of DNA epigenetic modification 5-hydroxymethylcytosine. Chem Sci 2025; 16:3953-3963. [PMID: 39906385 PMCID: PMC11788818 DOI: 10.1039/d4sc08660k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
Epigenetic modifications in genomes play a crucial role in regulating gene expression in mammals. Among these modifications, 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) are recognized as the fifth and sixth nucleobases in genomes, respectively, and are the two most significant epigenetic marks in mammals. 5hmC serves as both an intermediate in active DNA demethylation and a stable epigenetic modification involved in various biological processes. Analyzing the location of 5hmC is essential for understanding its functions. In this study, we introduce an orthologous mammalian A3A-mediated sequencing (OMA-seq) method for the quantitative detection of 5hmC in genomic DNA at single-nucleotide resolution. OMA-seq relies on the deamination properties of two naturally occurring mammalian A3A proteins: green monkey A3A (gmA3A) and dog A3A (dogA3A). The combined use of gmA3A and dogA3A effectively deaminates cytosine (C) and 5mC, but not 5hmC. As a result, the original C and 5mC in DNA are deaminated and read as thymine (T) during sequencing, while the original 5hmC remains unchanged and is read as C. Consequently, the remaining C in the sequence indicates the presence of original 5hmC. Using OMA-seq, we successfully quantified 5hmC in genomic DNA from lung cancer tissue and corresponding normal tissue. OMA-seq enables accurate and quantitative mapping of 5hmC at single-nucleotide resolution, utilizing a pioneering single-step deamination protocol that leverages the high specificity of natural deaminases. This approach eliminates the need for bisulfite conversion, DNA glycosylation, chemical oxidation, or screening of engineered protein variants, thereby streamlining the analysis of 5hmC. The utilization of orthologous enzymes for 5hmC detection expands the toolkit for epigenetic research, enabling the precise mapping of modified nucleosides and uncovering new insights into epigenetic regulation.
Collapse
Affiliation(s)
- Xia Guo
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
- Research Center of Public Health, Renmin Hospital of Wuhan University Wuhan 430060 China
- College of Chemistry and Molecular Sciences, Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, Wuhan University Wuhan 430072 China
| | - Jianyuan Wu
- Clinical Trial Center, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Tong-Tong Ji
- College of Chemistry and Molecular Sciences, Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, Wuhan University Wuhan 430072 China
| | - Min Wang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Shan Zhang
- College of Chemistry and Molecular Sciences, Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, Wuhan University Wuhan 430072 China
| | - Jun Xiong
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Fang-Yin Gang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Wei Liu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Yao-Hua Gu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
- School of Nursing, Wuhan University Wuhan 430071 China
| | - Yu Liu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
- Hubei Key Laboratory of Tumor Biological Behaviors, Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Neng-Bin Xie
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
| | - Bi-Feng Yuan
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University Wuhan 430071 China
- Research Center of Public Health, Renmin Hospital of Wuhan University Wuhan 430060 China
- College of Chemistry and Molecular Sciences, Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, Wuhan University Wuhan 430072 China
| |
Collapse
|
6
|
Qian C, Lee Y, Han Y, Zhong Y, Zhou J, Hrit J, Xie L, Chen Q, Kaniskan HÜ, Chen X, Rothbart S, Cheng X, Xiong Y, Jin J. Structure-Activity Relationship Studies of DNA Methyltransferase 1 Monovalent Degraders. J Med Chem 2025; 68:2903-2919. [PMID: 39905966 PMCID: PMC11932022 DOI: 10.1021/acs.jmedchem.4c02161] [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] [Indexed: 02/06/2025]
Abstract
DNA methyltransferase 1 (DNMT1), which catalyzes maintenance methylation of hemimethylated DNA during DNA replication, is overexpressed in cancer. Recently, the first-in-class DNMT1-selective noncovalent small-molecule inhibitors, GSK3484862 and GSK3685032, were discovered. These inhibitors were also reported to degrade DNMT1. However, structure-activity relationship (SAR) studies of these monovalent DNMT1 degraders are lacking. Here, we report our SAR studies of this scaffold on degrading DNMT1, which led to the discovery of multiple lead degraders, including compound 4 (MS9024). Compound 4 potently and selectively degraded DNMT1 in multiple cancer cell lines in a concentration-, time-, and proteasome-dependent manner without altering DNMT1 transcription. Further mechanism-of-action studies suggest that the DNMT1 degradation induced by 4 was not mediated by lysosome or cullin RING E3 ligases but could potentially be mediated by HECT E3 ligases and/or UHRF1. Collectively, these studies pave the way for further developing DNMT1 monovalent degraders as potential therapeutics and useful chemical tools.
Collapse
Affiliation(s)
- Chao Qian
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Youngeun Lee
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Yulin Han
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Yue Zhong
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jujun Zhou
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Joel Hrit
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan 49503, United States
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Qin Chen
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - H Ümit Kaniskan
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Scott Rothbart
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan 49503, United States
| | - Xiaodong Cheng
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
7
|
Chen X, Xu H, Shu X, Song CX. Mapping epigenetic modifications by sequencing technologies. Cell Death Differ 2025; 32:56-65. [PMID: 37658169 PMCID: PMC11742697 DOI: 10.1038/s41418-023-01213-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 09/03/2023] Open
Abstract
The "epigenetics" concept was first described in 1942. Thus far, chemical modifications on histones, DNA, and RNA have emerged as three important building blocks of epigenetic modifications. Many epigenetic modifications have been intensively studied and found to be involved in most essential biological processes as well as human diseases, including cancer. Precisely and quantitatively mapping over 100 [1], 17 [2], and 160 [3] different known types of epigenetic modifications in histone, DNA, and RNA is the key to understanding the role of epigenetic modifications in gene regulation in diverse biological processes. With the rapid development of sequencing technologies, scientists are able to detect specific epigenetic modifications with various quantitative, high-resolution, whole-genome/transcriptome approaches. Here, we summarize recent advances in epigenetic modification sequencing technologies, focusing on major histone, DNA, and RNA modifications in mammalian cells.
Collapse
Affiliation(s)
- Xiufei Chen
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Haiqi Xu
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Xiao Shu
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Chun-Xiao Song
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
| |
Collapse
|
8
|
Wang N, Liu Q, Wang B, Yang Z, Li S, Li R, Liang X, Fan J, Wang H, Sun Z, Dong L, Hou Y, Wang S, Song C, Wang Y, Quan C, Yang Q, Wang L. Host RNA N 6-methyladenosine and incoming DNA N 6-methyldeoxyadenosine modifications cooperatively elevate the condensation potential of DNA to activate immune surveillance. Mol Ther 2024; 32:4418-4434. [PMID: 39473181 PMCID: PMC11638879 DOI: 10.1016/j.ymthe.2024.10.027] [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: 03/27/2024] [Revised: 09/03/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Self-non-self discrimination is fundamental to life, thereby even microbes can apply DNA modifications to recognize non-self DNA. However, mammalian cytosolic DNA sensors indiscriminately bind DNA, necessitating specific mechanism(s) for self-non-self discrimination. Here, we show that mammalian RNA N6-methyladenosine (m6A) and incoming DNA N6-methyldeoxyadenosine (6mdA) cooperatively elevate the condensation potential of DNA to activate immunosurveillance. RNA m6A modification was found to enhance the activation of cyclic guanosine monophosphate-AMP synthase (cGAS) via increasing DNA phase separation. And 6mdA further increased the phase separation potential of DNA. Consistently, host RNA m6A and incoming DNA 6mdA modifications cooperatively elevated the incoming DNA condensation and cGAS activation. Moreover, we developed a prodrug, QKY-613. QKY-613 promoted a discriminative incorporation of 6mdA into viral DNAs to elevate host immune surveillance, and decreased mortality in virus-infected aged mice. Our results link nucleic acid modification diversity with immune surveillance via phase separation, which might be targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Na Wang
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Qiaoling Liu
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Bo Wang
- Department of General Surgery, The Second Affiliated Hospital of DaLian Medical University, Dalian, China
| | - Zhuo Yang
- Department of Gynecology, Cancer Hospital of DaLian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Siru Li
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Ran Li
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Xinyuan Liang
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Jiayu Fan
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Hui Wang
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Zhen Sun
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Ling Dong
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Yueru Hou
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Shengnan Wang
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Chengli Song
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Yang Wang
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China
| | - Chunshan Quan
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, China
| | - Qingkai Yang
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China.
| | - Lina Wang
- Institute of Cancer Stem Cell, DaLian Medical University, Dalian 116044, China.
| |
Collapse
|
9
|
Xu GE, Yu P, Hu Y, Wan W, Shen K, Cui X, Wang J, Wang T, Cui C, Chatterjee E, Li G, Cretoiu D, Sluijter JPG, Xu J, Wang L, Xiao J. Exercise training decreases lactylation and prevents myocardial ischemia-reperfusion injury by inhibiting YTHDF2. Basic Res Cardiol 2024; 119:651-671. [PMID: 38563985 DOI: 10.1007/s00395-024-01044-2] [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: 08/12/2023] [Revised: 02/19/2024] [Accepted: 03/02/2024] [Indexed: 04/04/2024]
Abstract
Exercise improves cardiac function and metabolism. Although long-term exercise leads to circulating and micro-environmental metabolic changes, the effect of exercise on protein post-translational lactylation modifications as well as its functional relevance is unclear. Here, we report that lactate can regulate cardiomyocyte changes by improving protein lactylation levels and elevating intracellular N6-methyladenosine RNA-binding protein YTHDF2. The intrinsic disorder region of YTHDF2 but not the RNA m6A-binding activity is indispensable for its regulatory function in influencing cardiomyocyte cell size changes and oxygen glucose deprivation/re-oxygenation (OGD/R)-stimulated apoptosis via upregulating Ras GTPase-activating protein-binding protein 1 (G3BP1). Downregulation of YTHDF2 is required for exercise-induced physiological cardiac hypertrophy. Moreover, myocardial YTHDF2 inhibition alleviated ischemia/reperfusion-induced acute injury and pathological remodeling. Our results here link lactate and lactylation modifications with RNA m6A reader YTHDF2 and highlight the physiological importance of this innovative post-transcriptional intrinsic regulation mechanism of cardiomyocyte responses to exercise. Decreasing lactylation or inhibiting YTHDF2/G3BP1 might represent a promising therapeutic strategy for cardiac diseases.
Collapse
Affiliation(s)
- Gui-E Xu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Life Science, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Pujiao Yu
- Department of Cardiology, Shanghai Gongli Hospital, Shanghai, 200135, China
| | - Yuxue Hu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Life Science, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Wensi Wan
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Life Science, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Keting Shen
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Life Science, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xinxin Cui
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Life Science, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiaqi Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Life Science, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Tianhui Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Life Science, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Caiyue Cui
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Dragos Cretoiu
- Department of Medical Genetics, Carol Davila University of Medicine and Pharmacy, 020031, Bucharest, Romania
- Materno-Fetal Assistance Excellence Unit, Alessandrescu-Rusescu National Institute for Mother and Child Health, 011062, Bucharest, Romania
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3508GA, Utrecht, The Netherlands
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Utrecht, 3508GA, The Netherlands
| | - Jiahong Xu
- Department of Cardiology, Shanghai Gongli Hospital, Shanghai, 200135, China.
| | - Lijun Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Life Science, Shanghai University, Nantong, 226011, China.
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China.
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Life Science, Shanghai University, Nantong, 226011, China.
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
10
|
Lax C, Mondo SJ, Osorio-Concepción M, Muszewska A, Corrochano-Luque M, Gutiérrez G, Riley R, Lipzen A, Guo J, Hundley H, Amirebrahimi M, Ng V, Lorenzo-Gutiérrez D, Binder U, Yang J, Song Y, Cánovas D, Navarro E, Freitag M, Gabaldón T, Grigoriev IV, Corrochano LM, Nicolás FE, Garre V. Symmetric and asymmetric DNA N6-adenine methylation regulates different biological responses in Mucorales. Nat Commun 2024; 15:6066. [PMID: 39025853 PMCID: PMC11258239 DOI: 10.1038/s41467-024-50365-2] [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] [Accepted: 07/05/2024] [Indexed: 07/20/2024] Open
Abstract
DNA N6-adenine methylation (6mA) has recently gained importance as an epigenetic modification in eukaryotes. Its function in lineages with high levels, such as early-diverging fungi (EDF), is of particular interest. Here, we investigated the biological significance and evolutionary implications of 6mA in EDF, which exhibit divergent evolutionary patterns in 6mA usage. The analysis of two Mucorales species displaying extreme 6mA usage reveals that species with high 6mA levels show symmetric methylation enriched in highly expressed genes. In contrast, species with low 6mA levels show mostly asymmetric 6mA. Interestingly, transcriptomic regulation throughout development and in response to environmental cues is associated with changes in the 6mA landscape. Furthermore, we identify an EDF-specific methyltransferase, likely originated from endosymbiotic bacteria, as responsible for asymmetric methylation, while an MTA-70 methylation complex performs symmetric methylation. The distinct phenotypes observed in the corresponding mutants reinforced the critical role of both types of 6mA in EDF.
Collapse
Affiliation(s)
- Carlos Lax
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Stephen J Mondo
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
- Department of Agricultural Biology, Colorado State University, Fort Collins, CO, 80523, USA
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Macario Osorio-Concepción
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Anna Muszewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106, Warsaw, Poland
| | | | - Gabriel Gutiérrez
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Robert Riley
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Anna Lipzen
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Jie Guo
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Hope Hundley
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Mojgan Amirebrahimi
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Vivian Ng
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Damaris Lorenzo-Gutiérrez
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Ulrike Binder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Junhuan Yang
- College of Food Science and Engineering, Lingnan Normal University, Zhanjiang, 524048, China
| | - Yuanda Song
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255049, China
| | - David Cánovas
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Eusebio Navarro
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Michael Freitag
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, 97331, USA
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Igor V Grigoriev
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
- Department of Plant and Microbial Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Luis M Corrochano
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain.
| | - Francisco E Nicolás
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain.
| | - Victoriano Garre
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain.
| |
Collapse
|
11
|
Sheng Y, Wang Y, Yang W, Wang XQ, Lu J, Pan B, Nan B, Liu Y, Ye F, Li C, Song J, Dou Y, Gao S, Liu Y. Semiconservative transmission of DNA N 6-adenine methylation in a unicellular eukaryote. Genome Res 2024; 34:740-756. [PMID: 38744529 PMCID: PMC11216311 DOI: 10.1101/gr.277843.123] [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: 03/03/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024]
Abstract
Although DNA N 6-adenine methylation (6mA) is best known in prokaryotes, its presence in eukaryotes has recently generated great interest. Biochemical and genetic evidence supports that AMT1, an MT-A70 family methyltransferase (MTase), is crucial for 6mA deposition in unicellular eukaryotes. Nonetheless, the 6mA transmission mechanism remains to be elucidated. Taking advantage of single-molecule real-time circular consensus sequencing (SMRT CCS), here we provide definitive evidence for semiconservative transmission of 6mA in Tetrahymena thermophila In wild-type (WT) cells, 6mA occurs at the self-complementary ApT dinucleotide, mostly in full methylation (full-6mApT); after DNA replication, hemi-methylation (hemi-6mApT) is transiently present on the parental strand, opposite to the daughter strand readily labeled by 5-bromo-2'-deoxyuridine (BrdU). In ΔAMT1 cells, 6mA predominantly occurs as hemi-6mApT. Hemi-to-full conversion in WT cells is fast, robust, and processive, whereas de novo methylation in ΔAMT1 cells is slow and sporadic. In Tetrahymena, regularly spaced 6mA clusters coincide with the linker DNA of nucleosomes arrayed in the gene body. Importantly, in vitro methylation of human chromatin by the reconstituted AMT1 complex recapitulates preferential targeting of hemi-6mApT sites in linker DNA, supporting AMT1's intrinsic and autonomous role in maintenance methylation. We conclude that 6mA is transmitted by a semiconservative mechanism: full-6mApT is split by DNA replication into hemi-6mApT, which is restored to full-6mApT by AMT1-dependent maintenance methylation. Our study dissects AMT1-dependent maintenance methylation and AMT1-independent de novo methylation, reveals a 6mA transmission pathway with a striking similarity to 5-methylcytosine (5mC) transmission at the CpG dinucleotide, and establishes 6mA as a bona fide eukaryotic epigenetic mark.
Collapse
Affiliation(s)
- Yalan Sheng
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yuanyuan Wang
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Wentao Yang
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | - Xue Qing Wang
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | - Jiuwei Lu
- Department of Biochemistry, University of California Riverside, Riverside, California 92521, USA
| | - Bo Pan
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Bei Nan
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yongqiang Liu
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Fei Ye
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Chun Li
- Division of Biostatistics, Department of Preventive Medicine, University of Southern California, Los Angeles, California 90033, USA
| | - Jikui Song
- Department of Biochemistry, University of California Riverside, Riverside, California 92521, USA
| | - Yali Dou
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | - Shan Gao
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yifan Liu
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA;
| |
Collapse
|
12
|
Chen S, Lai W, Wang H. Recent advances in high-performance liquid chromatography tandem mass spectrometry techniques for analysis of DNA damage and epigenetic modifications. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2024; 896:503755. [PMID: 38821674 DOI: 10.1016/j.mrgentox.2024.503755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 06/02/2024]
Abstract
Environmental exposure would cause DNA damage and epigenetic modification changes, potentially resulting in physiological dysfunction, thereby triggering diseases and even cancer. DNA damage and epigenetic modifications are thus promising biomarkers for environmental exposures and disease states. Benefiting from its high sensitivity and accuracy, high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) is considered the "gold standard technique" for investigating epigenetic DNA modifications. This review summarizes the recent advancements of UHPLC-MS/MS-based technologies for DNA damage and epigenetic modifications analysis, mainly focusing on the innovative methods developed for UHPLC-MS/MS-related pretreatment technologies containing efficient genomic DNA digestion and effective removal of the inorganic salt matrix, and the new strategies for improving detection sensitivity of liquid chromatography-mass spectrometry. Moreover, we also summarized the novel hyphenated techniques of the advanced UHPLC-MS/MS coupled with other separation and analysis methods for the measurement of DNA damage and epigenetic modification changes in special regions and fragments of chromosomes.
Collapse
Affiliation(s)
- Shaokun Chen
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Weiyi Lai
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Hailin Wang
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Institute of Environment and Health, Institute for Advanced Study, UCAS, Hangzhou 310000, China
| |
Collapse
|
13
|
Zhang L, Mu Y, Li T, Hu J, Lin H, Zhang L. Molecular basis of an atypical dsDNA 5mC/6mA bifunctional dioxygenase CcTet from Coprinopsis cinerea in catalyzing dsDNA 5mC demethylation. Nucleic Acids Res 2024; 52:3886-3895. [PMID: 38324471 PMCID: PMC11040006 DOI: 10.1093/nar/gkae066] [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: 09/05/2023] [Revised: 11/22/2023] [Accepted: 01/24/2024] [Indexed: 02/09/2024] Open
Abstract
The eukaryotic epigenetic modifications 5-methyldeoxycytosine (5mC) and N6-methyldeoxyadenine (6mA) have indispensable regulatory roles in gene expression and embryonic development. We recently identified an atypical bifunctional dioxygenase CcTet from Coprinopsis cinerea that works on both 5mC and 6mA demethylation. The nonconserved residues Gly331 and Asp337 of CcTet facilitate 6mA accommodation, while D337F unexpectedly abolishes 5mC oxidation activity without interfering 6mA demethylation, indicating a prominent distinct but unclear 5mC oxidation mechanism to the conventional Tet enzymes. Here, we assessed the molecular mechanism of CcTet in catalyzing 5mC oxidation by representing the crystal structure of CcTet-5mC-dsDNA complex. We identified the distinct mechanism by which CcTet recognizes 5mC-dsDNA compared to 6mA-dsDNA substrate. Moreover, Asp337 was found to have a central role in compensating for the loss of a critical 5mC-stablizing H-bond observed in conventional Tet enzymes, and stabilizes 5mC and subsequent intermediates through an H-bond with the N4 atom of the substrates. These findings improve our understanding of Tet enzyme functions in the dsDNA 5mC and 6mA demethylation pathways, and provide useful information for future discovery of small molecular probes targeting Tet enzymes in DNA active demethylation processes.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yajuan Mu
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tingting Li
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jingyan Hu
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Houwen Lin
- Research Centre for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Institute of Marine Biomedicine, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Liang Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
14
|
Meng WY, Wang ZX, Zhang Y, Hou Y, Xue JH. Epigenetic marks or not? The discovery of novel DNA modifications in eukaryotes. J Biol Chem 2024; 300:106791. [PMID: 38403247 PMCID: PMC11065753 DOI: 10.1016/j.jbc.2024.106791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/24/2024] [Accepted: 02/04/2024] [Indexed: 02/27/2024] Open
Abstract
DNA modifications add another layer of complexity to the eukaryotic genome to regulate gene expression, playing critical roles as epigenetic marks. In eukaryotes, the study of DNA epigenetic modifications has been confined to 5mC and its derivatives for decades. However, rapid developing approaches have witnessed the expansion of DNA modification reservoirs during the past several years, including the identification of 6mA, 5gmC, 4mC, and 4acC in diverse organisms. However, whether these DNA modifications function as epigenetic marks requires careful consideration. In this review, we try to present a panorama of all the DNA epigenetic modifications in eukaryotes, emphasizing recent breakthroughs in the identification of novel DNA modifications. The characterization of their roles in transcriptional regulation as potential epigenetic marks is summarized. More importantly, the pathways for generating or eliminating these DNA modifications, as well as the proteins involved are comprehensively dissected. Furthermore, we briefly discuss the potential challenges and perspectives, which should be taken into account while investigating novel DNA modifications.
Collapse
Affiliation(s)
- Wei-Ying Meng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zi-Xin Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yunfang Zhang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yujun Hou
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | - Jian-Huang Xue
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| |
Collapse
|
15
|
Liu Y, Niu J, Ye F, Solberg T, Lu B, Wang C, Nowacki M, Gao S. Dynamic DNA N 6-adenine methylation (6mA) governs the encystment process, showcased in the unicellular eukaryote Pseudocohnilembus persalinus. Genome Res 2024; 34:256-271. [PMID: 38471739 PMCID: PMC10984389 DOI: 10.1101/gr.278796.123] [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: 12/01/2023] [Accepted: 02/14/2024] [Indexed: 03/14/2024]
Abstract
The formation of resting cysts commonly found in unicellular eukaryotes is a complex and highly regulated survival strategy against environmental stress that involves drastic physiological and biochemical changes. Although most studies have focused on the morphology and structure of cysts, little is known about the molecular mechanisms that control this process. Recent studies indicate that DNA N 6-adenine methylation (6mA) could be dynamically changing in response to external stimuli; however, its potential role in the regulation of cyst formation remains unknown. We used the ciliate Pseudocohnilembus persalinus, which can be easily induced to form cysts to investigate the dynamic pattern of 6mA in trophonts and cysts. Single-molecule real-time (SMRT) sequencing reveals high levels of 6mA in trophonts that decrease in cysts, along with a conversion of symmetric 6mA to asymmetric 6mA. Further analysis shows that 6mA, a mark of active transcription, is involved in altering the expression of encystment-related genes through changes in 6mA levels and 6mA symmetric-to-asymmetric conversion. Most importantly, we show that reducing 6mA levels by knocking down the DNA 6mA methyltransferase PpAMT1 accelerates cyst formation. Taken together, we characterize the genome-wide 6mA landscape in P. persalinus and provide insights into the role of 6mA in gene regulation under environmental stress in eukaryotes. We propose that 6mA acts as a mark of active transcription to regulate the encystment process along with symmetric-to-asymmetric conversion, providing important information for understanding the molecular response to environmental cues from the perspective of 6mA modification.
Collapse
Affiliation(s)
- Yongqiang Liu
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Junhua Niu
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Fei Ye
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Therese Solberg
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
- Department of Molecular Biology, Keio University School of Medicine, 160-8582 Tokyo, Japan
- Human Biology Microbiome Quantum Research Center (WPI-Bio2Q), Keio University, 108-8345 Tokyo, Japan
| | - Borong Lu
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Chundi Wang
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory of Marine Protozoan Biodiversity and Evolution, Marine College, Shandong University, Weihai 264209, China
| | - Mariusz Nowacki
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | - Shan Gao
- MOE Key Laboratory of Evolution and Marine Biodiversity and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| |
Collapse
|
16
|
Feng X, Cui X, Zhang LS, Ye C, Wang P, Zhong Y, Wu T, Zheng Z, He C. Sequencing of N 6-methyl-deoxyadenosine at single-base resolution across the mammalian genome. Mol Cell 2024; 84:596-610.e6. [PMID: 38215754 PMCID: PMC10872247 DOI: 10.1016/j.molcel.2023.12.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 07/25/2023] [Accepted: 12/13/2023] [Indexed: 01/14/2024]
Abstract
Although DNA N6-methyl-deoxyadenosine (6mA) is abundant in bacteria and protists, its presence and function in mammalian genomes have been less clear. We present Direct-Read 6mA sequencing (DR-6mA-seq), an antibody-independent method, to measure 6mA at base resolution. DR-6mA-seq employs a unique mutation-based strategy to reveal 6mA sites as misincorporation signatures without any chemical or enzymatic modulation of 6mA. We validated DR-6mA-seq through the successful mapping of the well-characterized G(6mA)TC motif in the E. coli DNA. As expected, when applying DR-6mA-seq to mammalian systems, we found that genomic DNA (gDNA) 6mA abundance is generally low in most mammalian tissues and cells; however, we did observe distinct gDNA 6mA sites in mouse testis and glioblastoma cells. DR-6mA-seq provides an enabling tool to detect 6mA at single-base resolution for a comprehensive understanding of DNA 6mA in eukaryotes.
Collapse
Affiliation(s)
- Xinran Feng
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Xiaolong Cui
- Department of Chemistry, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Li-Sheng Zhang
- Department of Chemistry, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA; Department of Chemistry, Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Chang Ye
- Department of Chemistry, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Pingluan Wang
- Department of Chemistry, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Yuhao Zhong
- Department of Chemistry, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Tong Wu
- Department of Chemistry, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Zhong Zheng
- Department of Chemistry, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
17
|
Xie Y, Ruan F, Li Y, Luo M, Zhang C, Chen Z, Xie Z, Weng Z, Chen W, Chen W, Fang Y, Sun Y, Guo M, Wang J, Xu S, Wang H, Tang C. Spatial chromatin accessibility sequencing resolves high-order spatial interactions of epigenomic markers. eLife 2024; 12:RP87868. [PMID: 38236718 PMCID: PMC10945591 DOI: 10.7554/elife.87868] [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] [Indexed: 01/23/2024] Open
Abstract
As the genome is organized into a three-dimensional structure in intracellular space, epigenomic information also has a complex spatial arrangement. However, most epigenetic studies describe locations of methylation marks, chromatin accessibility regions, and histone modifications in the horizontal dimension. Proper spatial epigenomic information has rarely been obtained. In this study, we designed spatial chromatin accessibility sequencing (SCA-seq) to resolve the genome conformation by capturing the epigenetic information in single-molecular resolution while simultaneously resolving the genome conformation. Using SCA-seq, we are able to examine the spatial interaction of chromatin accessibility (e.g. enhancer-promoter contacts), CpG island methylation, and spatial insulating functions of the CCCTC-binding factor. We demonstrate that SCA-seq paves the way to explore the mechanism of epigenetic interactions and extends our knowledge in 3D packaging of DNA in the nucleus.
Collapse
Affiliation(s)
| | | | - Yaning Li
- BGI Genomics, BGI-ShenzhenShenzhenChina
| | - Meng Luo
- BGI Genomics, BGI-ShenzhenShenzhenChina
| | | | - Zhichao Chen
- BGI Genomics, BGI-ShenzhenShenzhenChina
- College of Life Sciences, University of Chinese Academy of SciencesBeijingChina
| | - Zhe Xie
- College of Life Sciences, University of Chinese Academy of SciencesBeijingChina
- Department of Biology, Cell Biology and Physiology, University of CopenhagenCopenhagenDenmark
| | - Zhe Weng
- BGI Genomics, BGI-ShenzhenShenzhenChina
| | - Weitian Chen
- BGI Genomics, BGI-ShenzhenShenzhenChina
- College of Life Sciences, University of Chinese Academy of SciencesBeijingChina
| | | | | | - Yuxin Sun
- BGI Genomics, BGI-ShenzhenShenzhenChina
| | - Mei Guo
- BGI Genomics, BGI-ShenzhenShenzhenChina
| | - Juan Wang
- BGI Genomics, BGI-ShenzhenShenzhenChina
| | - Shouping Xu
- Department of Breast Surgery, Harbin Medical University Cancer HospitalHarbinChina
| | | | | |
Collapse
|
18
|
Xie NB, Wang M, Chen W, Ji TT, Guo X, Gang FY, Wang YF, Feng YQ, Liang Y, Ci W, Yuan BF. Whole-Genome Sequencing of 5-Hydroxymethylcytosine at Base Resolution by Bisulfite-Free Single-Step Deamination with Engineered Cytosine Deaminase. ACS CENTRAL SCIENCE 2023; 9:2315-2325. [PMID: 38161361 PMCID: PMC10755730 DOI: 10.1021/acscentsci.3c01131] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
The epigenetic modification 5-hydroxymethylcytosine (5hmC) plays a crucial role in the regulation of gene expression. Although some methods have been developed to detect 5hmC, direct genome-wide mapping of 5hmC at base resolution is still highly desirable. Herein, we proposed a single-step deamination sequencing (SSD-seq) method, designed to precisely map 5hmC across the genome at single-base resolution. SSD-seq takes advantage of a screened engineered human apolipoprotein B mRNA-editing catalytic polypeptide-like 3A (A3A) protein, known as eA3A-v10, to selectively deaminate cytosine (C) and 5-methylcytosine (5mC) but not 5hmC. During sequencing, the deaminated C and 5mC are converted to uracil (U) and thymine (T), read as T in the sequencing data. However, 5hmC remains unaffected by eA3A-v10 and is read as C during sequencing. Consequently, the presence of C in the sequence reads indicates the original 5hmC. We applied SSD-seq to generate a base-resolution map of 5hmC in human lung tissue. Our findings revealed that 5hmC was predominantly localized to CpG dinucleotides. Furthermore, the base-resolution map of 5hmC generated by SSD-seq demonstrated a strong correlation with prior ACE-seq results. The advantages of SSD-seq are its single-step process, absence of bisulfite treatment or DNA glycosylation, cost effectiveness, and ability to detect and quantify 5hmC directly at single-base resolution.
Collapse
Affiliation(s)
- Neng-Bin Xie
- Department
of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
- Research
Center of Public Health, Renmin Hospital
of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Min Wang
- College
of Chemistry and Molecular Sciences, Wuhan
University, Wuhan 430072, China
| | - Wei Chen
- Department
of Laboratory Medicine, Zhongnan Hospital
of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Tong-Tong Ji
- College
of Chemistry and Molecular Sciences, Wuhan
University, Wuhan 430072, China
| | - Xia Guo
- College
of Chemistry and Molecular Sciences, Wuhan
University, Wuhan 430072, China
| | - Fang-Yin Gang
- Department
of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Ya-Feng Wang
- Department
of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Yu-Qi Feng
- Department
of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Yuan Liang
- Key
Laboratory of Genomics and Precision Medicine, and China National
Center for Bioinformation, Beijing Institute
of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Weimin Ci
- Key
Laboratory of Genomics and Precision Medicine, and China National
Center for Bioinformation, Beijing Institute
of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Bi-Feng Yuan
- Department
of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
- Research
Center of Public Health, Renmin Hospital
of Wuhan University, Wuhan University, Wuhan 430060, China
- College
of Chemistry and Molecular Sciences, Wuhan
University, Wuhan 430072, China
| |
Collapse
|
19
|
Boulet M, Gilbert G, Renaud Y, Schmidt-Dengler M, Plantié E, Bertrand R, Nan X, Jurkowski T, Helm M, Vandel L, Waltzer L. Adenine methylation is very scarce in the Drosophila genome and not erased by the ten-eleven translocation dioxygenase. eLife 2023; 12:RP91655. [PMID: 38126351 PMCID: PMC10735219 DOI: 10.7554/elife.91655] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
N6-methyladenine (6mA) DNA modification has recently been described in metazoans, including in Drosophila, for which the erasure of this epigenetic mark has been ascribed to the ten-eleven translocation (TET) enzyme. Here, we re-evaluated 6mA presence and TET impact on the Drosophila genome. Using axenic or conventional breeding conditions, we found traces of 6mA by LC-MS/MS and no significant increase in 6mA levels in the absence of TET, suggesting that this modification is present at very low levels in the Drosophila genome but not regulated by TET. Consistent with this latter hypothesis, further molecular and genetic analyses showed that TET does not demethylate 6mA but acts essentially in an enzymatic-independent manner. Our results call for further caution concerning the role and regulation of 6mA DNA modification in metazoans and underline the importance of TET non-enzymatic activity for fly development.
Collapse
Affiliation(s)
- Manon Boulet
- Université Clermont Auvergne, CNRS, INSERM, iGReDClermont-FerrandFrance
| | - Guerric Gilbert
- Université Clermont Auvergne, CNRS, INSERM, iGReDClermont-FerrandFrance
| | - Yoan Renaud
- Université Clermont Auvergne, CNRS, INSERM, iGReDClermont-FerrandFrance
| | - Martina Schmidt-Dengler
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-UniversitätMainzGermany
| | - Emilie Plantié
- Université Clermont Auvergne, CNRS, INSERM, iGReDClermont-FerrandFrance
| | - Romane Bertrand
- Université Clermont Auvergne, CNRS, INSERM, iGReDClermont-FerrandFrance
| | - Xinsheng Nan
- School of Biosciences, Cardiff UniversityCardiffUnited Kingdom
| | | | - Mark Helm
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-UniversitätMainzGermany
| | - Laurence Vandel
- Université Clermont Auvergne, CNRS, INSERM, iGReDClermont-FerrandFrance
| | - Lucas Waltzer
- Université Clermont Auvergne, CNRS, INSERM, iGReDClermont-FerrandFrance
| |
Collapse
|
20
|
Zhang J, Peng Q, Ma C, Wang J, Xiao C, Li T, Liu X, Zhou L, Xu X, Zhou WZ, Ding W, An NA, Zhang L, Liu Y, Li CY. 6mA-Sniper: Quantifying 6mA sites in eukaryotes at single-nucleotide resolution. SCIENCE ADVANCES 2023; 9:eadh7912. [PMID: 37862411 PMCID: PMC10588941 DOI: 10.1126/sciadv.adh7912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/18/2023] [Indexed: 10/22/2023]
Abstract
While N6-methyldeoxyadenine (6mA) modification is a fundamental regulation in prokaryotes, its prevalence and functions in eukaryotes are controversial. Here, we report 6mA-Sniper to quantify 6mA sites in eukaryotes at single-nucleotide resolution, and delineate a 6mA profile in Caenorhabditis elegans with 2034 sites. Twenty-six of 39 events with Mnl I restriction endonuclease sites were verified, demonstrating the feasibility of this method. The levels of 6mA sites pinpointed by 6mA-Sniper are generally increased after Pseudomonas aeruginosa infection, but decreased in strains with the removal of METL-9, the dominant 6mA methyltransferase. The enrichment of these sites on specific motif of [GC]GAG, the selective constrains on them, and their coordinated changes with METL-9 levels thus support an active shaping of the 6mA profile by methyltransferase. Moreover, for regions marked by 6mA sites that emerged after infection, an enrichment of up-regulated genes was detected, possibly mediated through a mutual exclusive cross-talk between 6mA and H3K27me3 modification. We thus highlight 6mA regulation as a previously neglected regulator in eukaryotes.
Collapse
Affiliation(s)
- Jie Zhang
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Qi Peng
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Chengchuan Ma
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Beijing Advanced Innovation Center for Genomics, Beijing 100871, China
| | - Jiaxin Wang
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Chunfu Xiao
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Ting Li
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Xiaoge Liu
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Liankui Zhou
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xinwei Xu
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Wei-Zhen Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqiu Ding
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Bioinformatics Core, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Ni A. An
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Li Zhang
- Chinese Institute for Brain Research, Beijing, China
| | - Ying Liu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Beijing Advanced Innovation Center for Genomics, Beijing 100871, China
| | - Chuan-Yun Li
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Southwest United Graduate School, Kunming 650092, China
| |
Collapse
|
21
|
Sturm Á, Sharma H, Bodnár F, Aslam M, Kovács T, Németh Á, Hotzi B, Billes V, Sigmond T, Tátrai K, Egyed B, Téglás-Huszár B, Schlosser G, Charmpilas N, Ploumi C, Perczel A, Tavernarakis N, Vellai T. N6-Methyladenine Progressively Accumulates in Mitochondrial DNA during Aging. Int J Mol Sci 2023; 24:14858. [PMID: 37834309 PMCID: PMC10573865 DOI: 10.3390/ijms241914858] [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: 08/04/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
N6-methyladenine (6mA) in the DNA is a conserved epigenetic mark with various cellular, physiological and developmental functions. Although the presence of 6mA was discovered a few years ago in the nuclear genome of distantly related animal taxa and just recently in mammalian mitochondrial DNA (mtDNA), accumulating evidence at present seriously questions the presence of N6-adenine methylation in these genetic systems, attributing it to methodological errors. In this paper, we present a reliable, PCR-based method to determine accurately the relative 6mA levels in the mtDNA of Caenorhabditis elegans, Drosophila melanogaster and dogs, and show that these levels gradually increase with age. Furthermore, daf-2(-)-mutant worms, which are defective for insulin/IGF-1 (insulin-like growth factor) signaling and live twice as long as the wild type, display a half rate at which 6mA progressively accumulates in the mtDNA as compared to normal values. Together, these results suggest a fundamental role for mtDNA N6-adenine methylation in aging and reveal an efficient diagnostic technique to determine age using DNA.
Collapse
Affiliation(s)
- Ádám Sturm
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
- Genetics Research Group, Eötvös Loránd Research Network-Eötvös Loránd University, 1117 Budapest, Hungary
| | - Himani Sharma
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
| | - Ferenc Bodnár
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
| | - Maryam Aslam
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
| | - Tibor Kovács
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
| | - Ákos Németh
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
| | - Bernadette Hotzi
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
- Genetics Research Group, Eötvös Loránd Research Network-Eötvös Loránd University, 1117 Budapest, Hungary
| | - Viktor Billes
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
- Genetics Research Group, Eötvös Loránd Research Network-Eötvös Loránd University, 1117 Budapest, Hungary
| | - Tímea Sigmond
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
| | - Kitti Tátrai
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
| | - Balázs Egyed
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
| | - Blanka Téglás-Huszár
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
| | - Gitta Schlosser
- Momentum Ion Mobility Mass Spectrometry Research Group, Hungarian Academy of Sciences-Eötvös Loránd University, 1117 Budapest, Hungary
| | - Nikolaos Charmpilas
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, P.O. Box 1385 Heraklion, Greece
| | - Christina Ploumi
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, P.O. Box 1385 Heraklion, Greece
| | - András Perczel
- Department of Organic Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, P.O. Box 1385 Heraklion, Greece
| | - Tibor Vellai
- Department of Genetics, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary; (H.S.); (B.E.)
- Genetics Research Group, Eötvös Loránd Research Network-Eötvös Loránd University, 1117 Budapest, Hungary
- Vellab Biotech Ltd., 6722 Szeged, Hungary
| |
Collapse
|
22
|
Ma CJ, Li G, Shao WX, Min YH, Wang P, Ding JH, Xie NB, Wang M, Tang F, Feng YQ, Ci W, Wang Y, Yuan BF. Single-Nucleotide Resolution Mapping of N6-Methyladenine in Genomic DNA. ACS CENTRAL SCIENCE 2023; 9:1799-1809. [PMID: 37780356 PMCID: PMC10540296 DOI: 10.1021/acscentsci.3c00481] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Indexed: 10/03/2023]
Abstract
N6-Methyladenine (6mA) is a naturally occurring DNA modification in both prokaryotes and eukaryotes. Herein, we developed a deaminase-mediated sequencing (DM-seq) method for genome-wide mapping of 6mA at single-nucleotide resolution. The method capitalizes on the selective deamination of adenine, but not 6mA, in DNA mediated by an evolved adenine deaminase, ABE8e. By employing this method, we achieved genome-wide mapping of 6mA in Escherichia coli and in mammalian mitochondrial DNA (mtDNA) at single-nucleotide resolution. We found that the 6mA sites are mainly located in the GATC motif in the E. coli genome. We also identified 17 6mA sites in mtDNA of HepG2 cells, where all of the 6mA sites are distributed in the heavy strand of mtDNA. We envision that DM-seq will be a valuable tool for uncovering new functions of 6mA in DNA and for exploring its potential roles in mitochondria-related human diseases.
Collapse
Affiliation(s)
- Cheng-Jie Ma
- School
of Public Health, Department of Radiation and Medical Oncology, Zhongnan
Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Sauvage
Center for Molecular Sciences, Department of Chemistry, Wuhan University, Wuhan 430072, China
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521-0403, United States
| | - Gaojie Li
- Key
Laboratory of Genomics and Precision Medicine, and China National
Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Xuan Shao
- School
of Public Health, Department of Radiation and Medical Oncology, Zhongnan
Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Sauvage
Center for Molecular Sciences, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Yi-Hao Min
- School
of Public Health, Department of Radiation and Medical Oncology, Zhongnan
Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Sauvage
Center for Molecular Sciences, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Ping Wang
- Key
Laboratory of Genomics and Precision Medicine, and China National
Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiang-Hui Ding
- School
of Public Health, Department of Radiation and Medical Oncology, Zhongnan
Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Sauvage
Center for Molecular Sciences, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Neng-Bin Xie
- School
of Public Health, Department of Radiation and Medical Oncology, Zhongnan
Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Sauvage
Center for Molecular Sciences, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Min Wang
- School
of Public Health, Department of Radiation and Medical Oncology, Zhongnan
Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Sauvage
Center for Molecular Sciences, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Feng Tang
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521-0403, United States
| | - Yu-Qi Feng
- School
of Public Health, Department of Radiation and Medical Oncology, Zhongnan
Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Sauvage
Center for Molecular Sciences, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Weimin Ci
- Key
Laboratory of Genomics and Precision Medicine, and China National
Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinsheng Wang
- Department
of Chemistry, University of California,
Riverside, Riverside, California 92521-0403, United States
| | - Bi-Feng Yuan
- School
of Public Health, Department of Radiation and Medical Oncology, Zhongnan
Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Sauvage
Center for Molecular Sciences, Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
23
|
Sturm Á, Saskői É, Hotzi B, Tarnóci A, Barna J, Bodnár F, Sharma H, Kovács T, Ari E, Weinhardt N, Kerepesi C, Perczel A, Ivics Z, Vellai T. Downregulation of transposable elements extends lifespan in Caenorhabditis elegans. Nat Commun 2023; 14:5278. [PMID: 37644049 PMCID: PMC10465613 DOI: 10.1038/s41467-023-40957-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/17/2023] [Indexed: 08/31/2023] Open
Abstract
Mobility of transposable elements (TEs) frequently leads to insertional mutations in functional DNA regions. In the potentially immortal germline, TEs are effectively suppressed by the Piwi-piRNA pathway. However, in the genomes of ageing somatic cells lacking the effects of the pathway, TEs become increasingly mobile during the adult lifespan, and their activity is associated with genomic instability. Whether the progressively increasing mobilization of TEs is a cause or a consequence of ageing remains a fundamental problem in biology. Here we show that in the nematode Caenorhabditis elegans, the downregulation of active TE families extends lifespan. Ectopic activation of Piwi proteins in the soma also promotes longevity. Furthermore, DNA N6-adenine methylation at TE stretches gradually rises with age, and this epigenetic modification elevates their transcription as the animal ages. These results indicate that TEs represent a novel genetic determinant of ageing, and that N6-adenine methylation plays a pivotal role in ageing control.
Collapse
Affiliation(s)
- Ádám Sturm
- Department of Genetics, Eötvös Loránd University (ELTE), 1117, Budapest, Hungary
- Eötvös Loránd Research Network (ELKH)-ELTE Genetics Research Group, 1117, Budapest, Hungary
| | - Éva Saskői
- Department of Genetics, Eötvös Loránd University (ELTE), 1117, Budapest, Hungary
| | - Bernadette Hotzi
- Department of Genetics, Eötvös Loránd University (ELTE), 1117, Budapest, Hungary
| | - Anna Tarnóci
- Eötvös Loránd Research Network (ELKH)-ELTE Genetics Research Group, 1117, Budapest, Hungary
| | - János Barna
- Eötvös Loránd Research Network (ELKH)-ELTE Genetics Research Group, 1117, Budapest, Hungary
| | - Ferenc Bodnár
- Department of Genetics, Eötvös Loránd University (ELTE), 1117, Budapest, Hungary
| | - Himani Sharma
- Department of Genetics, Eötvös Loránd University (ELTE), 1117, Budapest, Hungary
| | - Tibor Kovács
- Department of Genetics, Eötvös Loránd University (ELTE), 1117, Budapest, Hungary
| | - Eszter Ari
- Department of Genetics, Eötvös Loránd University (ELTE), 1117, Budapest, Hungary
- HCEMM-BRC Metabolic Systems Biology Research Group, 6726, Szeged, Hungary
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary
| | - Nóra Weinhardt
- Department of Genetics, Eötvös Loránd University (ELTE), 1117, Budapest, Hungary
| | - Csaba Kerepesi
- Institute for Computer Science and Control (SZTAKI), 1111, Budapest, Hungary
- Brigham and Women's Hospital & Harvard Medical School, Boston, MA, 02115, USA
| | - András Perczel
- Laboratory of Structural Chemistry and Biology & Hungarian Academy of Sciences (MTA)-ELTE Protein Modelling Research Group, Institute of Chemistry, Eötvös Loránd University, 1117, Budapest, Hungary
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, 63225, Langen, Germany
| | - Tibor Vellai
- Department of Genetics, Eötvös Loránd University (ELTE), 1117, Budapest, Hungary.
- Eötvös Loránd Research Network (ELKH)-ELTE Genetics Research Group, 1117, Budapest, Hungary.
- Vellab Biotech Ltd., 6722, Szeged, Hungary.
| |
Collapse
|
24
|
Febrimarsa, Gornik SG, Barreira SN, Salinas‐Saavedra M, Schnitzler CE, Baxevanis AD, Frank U. Randomly incorporated genomic N6-methyldeoxyadenosine delays zygotic transcription initiation in a cnidarian. EMBO J 2023; 42:e112934. [PMID: 37708295 PMCID: PMC10390872 DOI: 10.15252/embj.2022112934] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 09/16/2023] Open
Abstract
N6-methyldeoxyadenosine (6mA) is a chemical alteration of DNA, observed across all realms of life. Although the functions of 6mA are well understood in bacteria and protists, its roles in animal genomes have been controversial. We show that 6mA randomly accumulates in early embryos of the cnidarian Hydractinia symbiolongicarpus, with a peak at the 16-cell stage followed by clearance to background levels two cell cycles later, at the 64-cell stage-the embryonic stage at which zygotic genome activation occurs in this animal. Knocking down Alkbh1, a putative initiator of animal 6mA clearance, resulted in higher levels of 6mA at the 64-cell stage and a delay in the initiation of zygotic transcription. Our data are consistent with 6mA originating from recycled nucleotides of degraded m6A-marked maternal RNA postfertilization. Therefore, while 6mA does not function as an epigenetic mark in Hydractinia, its random incorporation into the early embryonic genome inhibits transcription. In turn, Alkbh1 functions as a genomic 6mA "cleaner," facilitating timely zygotic genome activation. Given the random nature of genomic 6mA accumulation and its ability to interfere with gene expression, defects in 6mA clearance may represent a hitherto unknown cause of various pathologies.
Collapse
Affiliation(s)
- Febrimarsa
- Centre for Chromosome Biology, School of Biological and Chemical SciencesUniversity of GalwayGalwayRepublic of Ireland
| | - Sebastian G Gornik
- Centre for Chromosome Biology, School of Biological and Chemical SciencesUniversity of GalwayGalwayRepublic of Ireland
- Present address:
Centre for Organismal StudiesHeidelberg UniversityHeidelbergGermany
| | - Sofia N Barreira
- Computational and Statistical Genomics Branch, Division of Intramural ResearchNational Human Genome Research Institute, National Institutes of HealthBethesdaMDUSA
| | - Miguel Salinas‐Saavedra
- Centre for Chromosome Biology, School of Biological and Chemical SciencesUniversity of GalwayGalwayRepublic of Ireland
| | - Christine E Schnitzler
- Whitney Laboratory for Marine BioscienceUniversity of FloridaSt. AugustineFLUSA
- Department of BiologyUniversity of FloridaGainesvilleFLUSA
| | - Andreas D Baxevanis
- Computational and Statistical Genomics Branch, Division of Intramural ResearchNational Human Genome Research Institute, National Institutes of HealthBethesdaMDUSA
| | - Uri Frank
- Centre for Chromosome Biology, School of Biological and Chemical SciencesUniversity of GalwayGalwayRepublic of Ireland
| |
Collapse
|
25
|
Ma C, Xue T, Peng Q, Zhang J, Guan J, Ding W, Li Y, Xia P, Zhou L, Zhao T, Wang S, Quan L, Li CY, Liu Y. A novel N 6-Deoxyadenine methyltransferase METL-9 modulates C. elegans immunity via dichotomous mechanisms. Cell Res 2023; 33:628-639. [PMID: 37271765 PMCID: PMC10397248 DOI: 10.1038/s41422-023-00826-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
N6-Methyldeoxyadenine (6mA) has been rediscovered as a DNA modification with potential biological function in metazoans. However, the physiological function and regulatory mechanisms regarding the establishment, maintenance and removal of 6mA in eukaryotes are still poorly understood. Here we show that genomic 6mA levels change in response to pathogenic infection in Caenorhabditis elegans (C. elegans). We further identify METL-9 as the methyltransferase that catalyzes DNA 6mA modifications upon pathogen infection. Deficiency of METL-9 impairs the induction of innate immune response genes and renders the animals more susceptible to pathogen infection. Interestingly, METL-9 functions through both 6mA-dependent and -independent mechanisms to transcriptionally regulate innate immunity. Our findings reveal that 6mA is a functional DNA modification in immunomodulation in C. elegans.
Collapse
Affiliation(s)
- Chengchuan Ma
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- Beijing Advanced Innovation Center for Genomics, Beijing, China.
- Research Center for Stem Cell and Regenerative Medicine, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China.
| | - Tingling Xue
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Qi Peng
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jie Zhang
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jialiang Guan
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- PKU-Tsinghua-NIBS Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Wanqiu Ding
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yi Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Peixue Xia
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Liankui Zhou
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Tianyu Zhao
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Sheng Wang
- Shanghai Zelixir Biotech Company Ltd., Shanghai, China
| | - Li Quan
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Chuan-Yun Li
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
| | - Ying Liu
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- Beijing Advanced Innovation Center for Genomics, Beijing, China.
| |
Collapse
|
26
|
Hu W, Guan L, Li M. Prediction of DNA Methylation based on Multi-dimensional feature encoding and double convolutional fully connected convolutional neural network. PLoS Comput Biol 2023; 19:e1011370. [PMID: 37639434 PMCID: PMC10461834 DOI: 10.1371/journal.pcbi.1011370] [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: 05/16/2023] [Accepted: 07/18/2023] [Indexed: 08/31/2023] Open
Abstract
DNA methylation takes on critical significance to the regulation of gene expression by affecting the stability of DNA and changing the structure of chromosomes. DNA methylation modification sites should be identified, which lays a solid basis for gaining more insights into their biological functions. Existing machine learning-based methods of predicting DNA methylation have not fully exploited the hidden multidimensional information in DNA gene sequences, such that the prediction accuracy of models is significantly limited. Besides, most models have been built in terms of a single methylation type. To address the above-mentioned issues, a deep learning-based method was proposed in this study for DNA methylation site prediction, termed the MEDCNN model. The MEDCNN model is capable of extracting feature information from gene sequences in three dimensions (i.e., positional information, biological information, and chemical information). Moreover, the proposed method employs a convolutional neural network model with double convolutional layers and double fully connected layers while iteratively updating the gradient descent algorithm using the cross-entropy loss function to increase the prediction accuracy of the model. Besides, the MEDCNN model can predict different types of DNA methylation sites. As indicated by the experimental results,the deep learning method based on coding from multiple dimensions outperformed single coding methods, and the MEDCNN model was highly applicable and outperformed existing models in predicting DNA methylation between different species. As revealed by the above-described findings, the MEDCNN model can be effective in predicting DNA methylation sites.
Collapse
Affiliation(s)
- Wenxing Hu
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Lixin Guan
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Mengshan Li
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, China
| |
Collapse
|
27
|
Chen S, Lai W, Li Y, Liu Y, Jiang J, Li X, Jiang G, Wang H. Aberrant DNA N 6 -methyladenine incorporation via adenylate kinase 1 is suppressed by ADAL deaminase-dependent 2'-deoxynucleotide pool sanitation. EMBO J 2023; 42:e113684. [PMID: 37366109 PMCID: PMC10390868 DOI: 10.15252/embj.2023113684] [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: 02/03/2023] [Revised: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
Intracellular decay of N6 -methyladenine (m6A)-containing RNA potentially induces aberrant N6 -methyl-2'-adenine (6mdA) misincorporation into DNA. Biophysically, misincorporated 6mdA may destabilize the DNA duplex in a manner similar to bona fide methylated 6mdA DNA, thereby affecting DNA replication and transcription. Utilizing heavy stable isotope labeling and ultrasensitive UHPLC-MS/MS assay, we demonstrate that intracellular m6A-RNA decay does not generate free 6mdA species, nor lead to any misincorporated DNA 6mdA in most mammalian cell lines tested, unveiling the existence of a sanitation mechanism that prevents 6mdA misincorporation. Depletion of deaminase ADAL increases the levels of free 6mdA species, concomitant with the presence of DNA-misincorporated 6mdA resulting from intracellular RNA m6A decay, suggesting that ADAL catabolizes 6mdAMP in vivo. Furthermore, we show that the overexpression of adenylate kinase 1 (AK1) promotes 6mdA misincorporation, while AK1 knockdown diminishes 6mdA incorporation, in ADAL-deficient cells. We conclude that ADAL together with other factors (such as MTH1) contributes to 2'-deoxynucleotide pool sanitation in most cells but compromised sanitation (e.g., in NIH3T3 cells) and increased AK1 expression may facilitate aberrant 6mdA incorporation. This sanitation mechanism may provide a framework for the maintenance of the epigenetic 6mdA landscape.
Collapse
Affiliation(s)
- Shaokun Chen
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijingChina
- School of Chemical SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Weiyi Lai
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijingChina
| | - Yanan Li
- Institute of Environment and Health, Institute for Advanced StudyUCASHangzhouChina
| | - Yan Liu
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijingChina
| | - Jie Jiang
- Shenzhen Center for Disease Control and PreventionShenzhenChina
| | - Xiangjun Li
- School of Chemical SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Guibin Jiang
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijingChina
- Institute of Environment and Health, Institute for Advanced StudyUCASHangzhouChina
| | - Hailin Wang
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijingChina
- Institute of Environment and Health, Institute for Advanced StudyUCASHangzhouChina
| |
Collapse
|
28
|
Kong Y, Mead EA, Fang G. Navigating the pitfalls of mapping DNA and RNA modifications. Nat Rev Genet 2023; 24:363-381. [PMID: 36653550 PMCID: PMC10722219 DOI: 10.1038/s41576-022-00559-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 01/19/2023]
Abstract
Chemical modifications to nucleic acids occur across the kingdoms of life and carry important regulatory information. Reliable high-resolution mapping of these modifications is the foundation of functional and mechanistic studies, and recent methodological advances based on next-generation sequencing and long-read sequencing platforms are critical to achieving this aim. However, mapping technologies may have limitations that sometimes lead to inconsistent results. Some of these limitations are technical in nature and specific to certain types of technology. Here, however, we focus on common (yet not always widely recognized) pitfalls that are shared among frequently used mapping technologies and discuss strategies to help technology developers and users mitigate their effects. Although the emphasis is primarily on DNA modifications, RNA modifications are also discussed.
Collapse
Affiliation(s)
- Yimeng Kong
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edward A Mead
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gang Fang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
29
|
Abstract
After decades of research, our knowledge of the complexity of cancer mechanisms, elegantly summarized as 'hallmarks of cancer', is expanding, as are the therapeutic opportunities that this knowledge brings. However, cancer still needs intense research to diminish its tremendous impact. In this context, the use of simple model organisms such as Caenorhabditis elegans, in which the genetics of the apoptotic pathway was discovered, can facilitate the investigation of several cancer hallmarks. Amenable for genetic and drug screens, convenient for fast and efficient genome editing, and aligned with the 3Rs ('Replacement, Reduction and Refinement') principles for ethical animal research, C. elegans plays a significant role in unravelling the intricate network of cancer mechanisms and presents a promising option in clinical diagnosis and drug discovery.
Collapse
Affiliation(s)
- Julián Cerón
- Modeling Human Diseases in C. elegans Group – Genes, Disease and Therapy Program, Bellvitge Biomedical Research Institute – IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
30
|
Pan B, Ye F, Li T, Wei F, Warren A, Wang Y, Gao S. Potential role of N 6-adenine DNA methylation in alternative splicing and endosymbiosis in Paramecium bursaria. iScience 2023; 26:106676. [PMID: 37182097 PMCID: PMC10173741 DOI: 10.1016/j.isci.2023.106676] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 12/02/2022] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
N6-adenine DNA methylation (6mA), a rediscovered epigenetic mark in eukaryotic organisms, diversifies in abundance, distribution, and function across species, necessitating its study in more taxa. Paramecium bursaria is a typical model organism with endosymbiotic algae of the species Chlorella variabilis. This consortium therefore serves as a valuable system to investigate the functional role of 6mA in endosymbiosis, as well as the evolutionary importance of 6mA among eukaryotes. In this study, we report the first genome-wide, base pair-resolution map of 6mA in P. bursaria and identify its methyltransferase PbAMT1. Functionally, 6mA exhibits a bimodal distribution at the 5' end of RNA polymerase II-transcribed genes and possibly participates in transcription by facilitating alternative splicing. Evolutionarily, 6mA co-evolves with gene age and likely serves as a reverse mark of endosymbiosis-related genes. Our results offer new insights for the functional diversification of 6mA in eukaryotes as an important epigenetic mark.
Collapse
Affiliation(s)
- Bo Pan
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao 266237, China
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Science, Ocean University of China, Qingdao 266003, China
| | - Fei Ye
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao 266237, China
| | - Tao Li
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao 266237, China
| | - Fan Wei
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao 266237, China
| | - Alan Warren
- Department of Life Sciences, Natural History Museum, London SW7 5BD, UK
| | - Yuanyuan Wang
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao 266237, China
- Corresponding author
| | - Shan Gao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao 266237, China
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Science, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
31
|
Voinea IC, Alistar CF, Banciu A, Popescu RG, Voicu SN, Nita-Lazar M, Vasile GG, Gheorghe S, Croitoru AM, Dolete G, Mihaiescu DE, Ficai A, Popa M, Marutescu L, Pircalabioru GG, Craciun N, Avramescu S, Marinescu GC, Chifiriuc MC, Stan MS, Dinischiotu A. Snapshot of the pollution-driven metabolic and microbiota changes in Carassius gibelio from Bucharest leisure lakes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 884:163810. [PMID: 37127150 DOI: 10.1016/j.scitotenv.2023.163810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
In the last decades, increased intakes of contaminants and the habitats' destruction have produced drastic changes in the aquatic ecosystems. The environmental contaminants can accumulate in aquatic organisms, leading to the disturbance of the antioxidant/prooxidant balance in fish. In this context, we evaluated the level of organic, inorganic and microbiological pollutants in four leisure lakes (Chitila, Floreasca, Tei and Vacaresti) from Bucharest, the largest city of Romania, in order to compare their effects on hepatopancreas and gills metabolism and antioxidant defense mechanisms in Carassius gibelio, the most known and widespread freshwater fish in this country. The lowest level of oxidative stress was recorded in the case of fish collected from the Vacaresti lake, a protected wetland area where aquatic organisms live in wild environmental conditions. In contrast, significant oxidative changes were observed in the hepatopancreas and gills of fish from the Chitila, Floreasca and Tei lakes, such as reduced glutathione S-transferase activity and glutathione level, and increased degree of lipid peroxidation, being correlated with elevated levels of pesticides (such as 2,4'-methoxychlor) and Escherichia coli load in these organs. Although different patterns of pollutants' accumulation were observed, no important interindividual variations in cytosine methylation degree were determined. In conclusion, the presence and concentrations of metals, pesticides and antibiotics varied with the analyzed tissue and sampling site, and were correlated with changes in the cellular redox homeostasis, but without significantly affecting the epigenetic mechanisms.
Collapse
Affiliation(s)
- Ionela C Voinea
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania; Research Institute of the University of Bucharest (ICUB), University of Bucharest, 050657 Bucharest, Romania
| | - Cristina F Alistar
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania
| | - Alina Banciu
- National Research and Development Institute for Industrial Ecology (ECOIND), 57-73 Drumul Podu Dambovitei, 060652 Bucharest, Romania
| | - Roua G Popescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania
| | - Sorina N Voicu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania
| | - Mihai Nita-Lazar
- National Research and Development Institute for Industrial Ecology (ECOIND), 57-73 Drumul Podu Dambovitei, 060652 Bucharest, Romania
| | - Gabriela Geanina Vasile
- National Research and Development Institute for Industrial Ecology (ECOIND), 57-73 Drumul Podu Dambovitei, 060652 Bucharest, Romania
| | - Stefania Gheorghe
- National Research and Development Institute for Industrial Ecology (ECOIND), 57-73 Drumul Podu Dambovitei, 060652 Bucharest, Romania
| | - Alexa-Maria Croitoru
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, Gh. Polizu St. 1-7, 060042 Bucharest, Romania; National Centre for Food Safety, University POLITEHNICA of Bucharest, Splaiul Independentei 313, Bucharest, Romania; National Centre for Micro- and Nanomaterials, University POLITEHNICA of Bucharest, Splaiul Independentei 313, Bucharest, Romania
| | - Georgiana Dolete
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, Gh. Polizu St. 1-7, 060042 Bucharest, Romania; National Centre for Food Safety, University POLITEHNICA of Bucharest, Splaiul Independentei 313, Bucharest, Romania; National Centre for Micro- and Nanomaterials, University POLITEHNICA of Bucharest, Splaiul Independentei 313, Bucharest, Romania
| | - Dan Eduard Mihaiescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, Gh. Polizu St. 1-7, 060042 Bucharest, Romania; National Centre for Food Safety, University POLITEHNICA of Bucharest, Splaiul Independentei 313, Bucharest, Romania; National Centre for Micro- and Nanomaterials, University POLITEHNICA of Bucharest, Splaiul Independentei 313, Bucharest, Romania; Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania
| | - Anton Ficai
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, Gh. Polizu St. 1-7, 060042 Bucharest, Romania; National Centre for Food Safety, University POLITEHNICA of Bucharest, Splaiul Independentei 313, Bucharest, Romania; National Centre for Micro- and Nanomaterials, University POLITEHNICA of Bucharest, Splaiul Independentei 313, Bucharest, Romania; Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania
| | - Marcela Popa
- Department of Microbiology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Luminita Marutescu
- Department of Microbiology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Gratiela Gradisteanu Pircalabioru
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, 050657 Bucharest, Romania; Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania; Department of Microbiology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Nicolae Craciun
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania
| | - Sorin Avramescu
- Department of Inorganic Chemistry, Organic Chemistry, Biochemistry and Catalysis, Faculty of Chemistry, University of Bucharest, 90-92 Soseaua Panduri, 050663 Bucharest, Romania; Research Center for Environmental Protection and Waste Management, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| | - George Catalin Marinescu
- Asociația Independent Research, 58 Timisului, 012416 Bucharest, Romania; Blue Screen SRL, 58 Timisului, 012416 Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, 050657 Bucharest, Romania; Department of Microbiology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania; The Romanian Academy, Calea Victoriei 25, District 1, 010071 Bucharest, Romania
| | - Miruna S Stan
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania; Research Institute of the University of Bucharest (ICUB), University of Bucharest, 050657 Bucharest, Romania.
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania
| |
Collapse
|
32
|
Lyu C, Wang HD, Lai W, Wang H. Identification and quantification of DNA N 6-methyladenine modification in mammals: A challenge to modern analytical technologies. Curr Opin Chem Biol 2023; 73:102259. [PMID: 36652775 DOI: 10.1016/j.cbpa.2022.102259] [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: 11/05/2022] [Revised: 12/08/2022] [Accepted: 12/11/2022] [Indexed: 01/18/2023]
Abstract
DNA N6-methyladenine modification (6mA) is a predominant epigenetic mark in prokaryotes but rarely present in multicellular metazoa. The analytical technologies have been developed for sensitive detection of 6mA, including ultra-high performance liquid chromatography coupled with mass spectrometry (UHPLC-MS/MS) and single molecule real-time sequencing (SMRTseq). However, it remains challenging to detect 6mA at global level and/or in the context of sequence in multicellular metazoa (including mammals). This mini-review brings insights into current dilemma and potential solutions for the identification and quantifications of 6mA in mammals.
Collapse
Affiliation(s)
- Cong Lyu
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Hui-Dong Wang
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Weiyi Lai
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Hailin Wang
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
33
|
Weng Z, Ruan F, Chen W, Chen Z, Xie Y, Luo M, Xie Z, Zhang C, Wang J, Sun Y, Fang Y, Guo M, Tan C, Chen W, Tong Y, Li Y, Wang H, Tang C. BIND&MODIFY: a long-range method for single-molecule mapping of chromatin modifications in eukaryotes. Genome Biol 2023; 24:61. [PMID: 36991510 PMCID: PMC10052867 DOI: 10.1186/s13059-023-02896-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Epigenetic modifications of histones are associated with development and pathogenesis of disease. Existing approaches cannot provide insights into long-range interactions and represent the average chromatin state. Here we describe BIND&MODIFY, a method using long-read sequencing for profiling histone modifications and transcription factors on individual DNA fibers. We use recombinant fused protein A-M.EcoGII to tether methyltransferase M.EcoGII to protein binding sites to label neighboring regions by methylation. Aggregated BIND&MODIFY signal matches bulk ChIP-seq and CUT&TAG. BIND&MODIFY can simultaneously measure histone modification status, transcription factor binding, and CpG 5mC methylation at single-molecule resolution and also quantifies correlation between local and distal elements.
Collapse
Affiliation(s)
- Zhe Weng
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | | | - Weitian Chen
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhichao Chen
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yeming Xie
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Meng Luo
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Zhe Xie
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
- Department of Biology, Cell Biology and Physiology, University of Copenhagen 13, 2100, Copenhagen, Denmark
| | - Chen Zhang
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Juan Wang
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Yuxin Sun
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Yitong Fang
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Mei Guo
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Chen Tan
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Wenfang Chen
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Yiqin Tong
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Yaning Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Hongqi Wang
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Chong Tang
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China.
| |
Collapse
|
34
|
Debo BM, Mallory BJ, Stergachis AB. Evaluation of N 6-methyldeoxyadenosine antibody-based genomic profiling in eukaryotes. Genome Res 2023; 33:427-434. [PMID: 36788024 PMCID: PMC10078290 DOI: 10.1101/gr.276696.122] [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: 03/02/2022] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Abstract
Low-level DNA N 6-methyldeoxyadenosine (DNA-m6A) has recently been reported across various eukaryotes. Although anti-m6A antibody-based approaches are commonly used to measure DNA-m6A levels, this approach is known to be confounded by DNA secondary structures, RNA contamination, and bacterial contamination. To evaluate for these confounding features, we introduce an approach for systematically validating the selectivity of antibody-based DNA-m6A methods and use a highly selective anti-DNA-m6A antibody to reexamine patterns of DNA-m6A in C. reinhardtii, A. thaliana, and D. melanogaster Our findings raise caution about the use of antibody-based methods for endogenous m6A quantification and mapping in eukaryotes.
Collapse
Affiliation(s)
- Brian M Debo
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Benjamin J Mallory
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA
| | - Andrew B Stergachis
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA;
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington 98195-7720, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington 98195, USA
| |
Collapse
|
35
|
Epigenetic Regulation of Ferroptosis in Central Nervous System Diseases. Mol Neurobiol 2023; 60:3584-3599. [PMID: 36847936 DOI: 10.1007/s12035-023-03267-1] [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: 08/29/2022] [Accepted: 02/14/2023] [Indexed: 03/01/2023]
Abstract
Ferroptosis, a newly identified form of cell death, is characterized by iron overload and accumulation of lipid reactive oxygen species. Inactivation of pathways, such as glutathione/glutathione peroxidase 4, NAD(P)H/ferroptosis suppressor protein 1/ubiquinone, dihydroorotate dehydrogenase/ubiquinol, or guanosine triphosphate cyclohydrolase-1/6(R)-L-erythro-5,6,7,8-tetrahydrobiopterin pathways, have been found to induce ferroptosis. The accumulating data suggest that epigenetic regulation can determine cell sensitivity to ferroptosis at both the transcriptional and translational levels. While many of the effectors that regulate ferroptosis have been mapped, epigenetic regulation in ferroptosis is not yet fully understood. Neuronal ferroptosis is a driver in several central nervous system (CNS) diseases, such as stroke, Parkinson's disease, traumatic brain injury, and spinal cord injury, and thus, research on how to inhibit neuronal ferroptosis is required to develop novel therapies for these diseases. In this review, we have summarized epigenetic regulation of ferroptosis in these CNS diseases, focusing in particular on DNA methylation, non-coding RNA regulation, and histone modification. Understanding epigenetic regulation in ferroptosis will hasten the development of promising therapeutic strategies in CNS diseases associated with ferroptosis.
Collapse
|
36
|
Feng X, He C. Mammalian DNA N 6-methyladenosine: Challenges and new insights. Mol Cell 2023; 83:343-351. [PMID: 36736309 PMCID: PMC10182828 DOI: 10.1016/j.molcel.2023.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 02/05/2023]
Abstract
DNA N6-methyldeoxyadenosine (6mA) modification was first discovered in Bacterium coli in the 1950s. Over the next several decades, 6mA was recognized as a critical DNA modification in the genomes of prokaryotes and protists. While important in prokaryotes, less is known about the presence and functional roles of DNA 6mA in eukaryotes, particularly in mammals. Taking advantage of recent technology advances that made 6mA detection and sequencing possible, studies over the past several years have brought new insights into 6mA biology in mammals. In this perspective, we present recent progress, discuss challenges, and pose four questions for future research regarding mammalian DNA 6mA.
Collapse
Affiliation(s)
- Xinran Feng
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
37
|
Broche J, Köhler AR, Kühnel F, Osteresch B, Chandrasekaran TT, Adam S, Brockmeyer J, Jeltsch A. Genome-wide deposition of 6-methyladenine in human DNA reduces the viability of HEK293 cells and directly influences gene expression. Commun Biol 2023; 6:138. [PMID: 36732350 PMCID: PMC9895073 DOI: 10.1038/s42003-023-04466-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
While cytosine-C5 methylation of DNA is an essential regulatory system in higher eukaryotes, the presence and relevance of 6-methyladenine (m6dA) in human cells is controversial. To study the role of m6dA in human DNA, we introduced it in human cells at a genome-wide scale at GANTC and GATC sites by expression of bacterial DNA methyltransferases and observed concomitant reductions in cell viability, in particular after global GANTC methylation. We identified several genes that are directly regulated by m6dA in a GANTC context. Upregulated genes showed m6dA-dependent reduction of H3K27me3 suggesting that the PRC2 complex is inhibited by m6dA. Genes downregulated by m6dA showed enrichment of JUN family transcription factor binding sites. JUN binds m6dA containing DNA with reduced affinity suggesting that m6dA can reduce the recruitment of JUN transcription factors to target genes. Our study documents that global introduction of m6dA in human DNA has physiological effects. Furthermore, we identified a set of target genes which are directly regulated by m6dA in human cells, and we defined two molecular pathways with opposing effects by which artificially introduced m6dA in GANTC motifs can directly control gene expression and phenotypes of human cells.
Collapse
Affiliation(s)
- Julian Broche
- Institute of Biochemistry and Technical Biochemistry, Department of Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Anja R Köhler
- Institute of Biochemistry and Technical Biochemistry, Department of Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Fiona Kühnel
- Institute of Biochemistry and Technical Biochemistry, Department of Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Bernd Osteresch
- Institute of Biochemistry and Technical Biochemistry, Department of Food Chemistry, University of Stuttgart, Allmandring 5b, 70569, Stuttgart, Germany
| | - Thyagarajan T Chandrasekaran
- Institute of Biochemistry and Technical Biochemistry, Department of Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Sabrina Adam
- Institute of Biochemistry and Technical Biochemistry, Department of Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Jens Brockmeyer
- Institute of Biochemistry and Technical Biochemistry, Department of Food Chemistry, University of Stuttgart, Allmandring 5b, 70569, Stuttgart, Germany
| | - Albert Jeltsch
- Institute of Biochemistry and Technical Biochemistry, Department of Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
| |
Collapse
|
38
|
Enam SU, Cherry JL, Leonard SR, Zheludev IN, Lipman DJ, Fire AZ. Restriction Endonuclease-Based Modification-Dependent Enrichment (REMoDE) of DNA for Metagenomic Sequencing. Appl Environ Microbiol 2023; 89:e0167022. [PMID: 36519847 PMCID: PMC9888230 DOI: 10.1128/aem.01670-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Metagenomic sequencing is a swift and powerful tool to ascertain the presence of an organism of interest in a sample. However, sequencing coverage of the organism of interest can be insufficient due to an inundation of reads from irrelevant organisms in the sample. Here, we report a nuclease-based approach to rapidly enrich for DNA from certain organisms, including enterobacteria, based on their differential endogenous modification patterns. We exploit the ability of taxon-specific methylated motifs to resist the action of cognate methylation-sensitive restriction endonucleases that thereby digest unwanted, unmethylated DNA. Subsequently, we use a distributive exonuclease or electrophoretic separation to deplete or exclude the digested fragments, thus enriching for undigested DNA from the organism of interest. As a proof of concept, we apply this method to enrich for the enterobacteria Escherichia coli and Salmonella enterica by 11- to 142-fold from mock metagenomic samples and validate this approach as a versatile means to enrich for genomes of interest in metagenomic samples. IMPORTANCE Pathogens that contaminate the food supply or spread through other means can cause outbreaks that bring devastating repercussions to the health of a populace. Investigations to trace the source of these outbreaks are initiated rapidly but can be drawn out due to the labored methods of pathogen isolation. Metagenomic sequencing can alleviate this hurdle but is often insufficiently sensitive. The approach and implementations detailed here provide a rapid means to enrich for many pathogens involved in foodborne outbreaks, thereby improving the utility of metagenomic sequencing as a tool in outbreak investigations. Additionally, this approach provides a means to broadly enrich for otherwise minute levels of modified DNA, which may escape unnoticed in metagenomic samples.
Collapse
Affiliation(s)
- Syed Usman Enam
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Joshua L. Cherry
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Susan R. Leonard
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, USA
| | - Ivan N. Zheludev
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, USA
| | - David J. Lipman
- Office of the Center Director, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Andrew Z. Fire
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
39
|
Yang M, Leng D, Zeng B, Wang T, Xu Z, Li D. Characteristics and functions of DNA N(6)-methyladenine in embryonic chicken muscle development. Poult Sci 2023; 102:102528. [PMID: 36907131 PMCID: PMC10024188 DOI: 10.1016/j.psj.2023.102528] [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: 11/11/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
DNA N(6)-methyladenine (DNA-6mA) is a new epigenetic mark in eukaryotes, the distribution and functions of which in genomic DNA remain unknown. Although recent studies have suggested that 6mA is present in multiple model organisms and is dynamically regulated during development, the genomic features of 6mA in avian species have yet to be elucidated. 6mA immunoprecipitation sequencing approach was used to analysis the distribution and function of 6mA in the muscle genomic DNA during embryonic chicken development. 6mA immunoprecipitation sequencing was combined with transcriptomic sequencing to reveal the role of 6mA in the regulation of gene expression and to explore possible pathways by which 6mA is involved in muscle development. We here provide evidence that 6mA modification exists widely throughout the chicken genome, and show preliminary data regarding genome-wide distribution of this epigenetic mark. Gene expression was shown to be inhibited by 6mA modification in promoter regions. In addition, the promoters of some genes related to development were modified by 6mA, indicating that 6mA may be involved in embryonic chicken development. Furthermore, 6mA may participate in muscle development and immune function by regulating HSPB8 and OASL expression. Our study improves our understanding of the distribution and function of 6mA modification in higher organisms and provide new information about differences between mammals and other vertebrates. These findings demonstrate an epigenetic role for 6mA in gene expression and potential involvement in chicken muscle development. Furthermore, the results suggest a potential epigenetic role for 6mA in avian embryonic development.
Collapse
Affiliation(s)
- Maosen Yang
- School of Pharmacy, Chengdu University, Chengdu 610106, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Dong Leng
- School of Pharmacy, Chengdu University, Chengdu 610106, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Bo Zeng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Wang
- School of Pharmacy, Chengdu University, Chengdu 610106, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhongxian Xu
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong 637002, China
| | - Diyan Li
- School of Pharmacy, Chengdu University, Chengdu 610106, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
40
|
Nabeel Asim M, Ali Ibrahim M, Fazeel A, Dengel A, Ahmed S. DNA-MP: a generalized DNA modifications predictor for multiple species based on powerful sequence encoding method. Brief Bioinform 2023; 24:6931721. [PMID: 36528802 DOI: 10.1093/bib/bbac546] [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: 06/14/2022] [Revised: 11/06/2022] [Accepted: 11/12/2022] [Indexed: 12/23/2022] Open
Abstract
Accurate prediction of deoxyribonucleic acid (DNA) modifications is essential to explore and discern the process of cell differentiation, gene expression and epigenetic regulation. Several computational approaches have been proposed for particular type-specific DNA modification prediction. Two recent generalized computational predictors are capable of detecting three different types of DNA modifications; however, type-specific and generalized modifications predictors produce limited performance across multiple species mainly due to the use of ineffective sequence encoding methods. The paper in hand presents a generalized computational approach "DNA-MP" that is competent to more precisely predict three different DNA modifications across multiple species. Proposed DNA-MP approach makes use of a powerful encoding method "position specific nucleotides occurrence based 117 on modification and non-modification class densities normalized difference" (POCD-ND) to generate the statistical representations of DNA sequences and a deep forest classifier for modifications prediction. POCD-ND encoder generates statistical representations by extracting position specific distributional information of nucleotides in the DNA sequences. We perform a comprehensive intrinsic and extrinsic evaluation of the proposed encoder and compare its performance with 32 most widely used encoding methods on $17$ benchmark DNA modifications prediction datasets of $12$ different species using $10$ different machine learning classifiers. Overall, with all classifiers, the proposed POCD-ND encoder outperforms existing $32$ different encoders. Furthermore, combinedly over 5-fold cross validation benchmark datasets and independent test sets, proposed DNA-MP predictor outperforms state-of-the-art type-specific and generalized modifications predictors by an average accuracy of 7% across 4mc datasets, 1.35% across 5hmc datasets and 10% for 6ma datasets. To facilitate the scientific community, the DNA-MP web application is available at https://sds_genetic_analysis.opendfki.de/DNA_Modifications/.
Collapse
Affiliation(s)
- Muhammad Nabeel Asim
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern 67663, Germany.,German Research Center for Artificial Intelligence GmbH, Kaiserslautern 67663, Germany
| | - Muhammad Ali Ibrahim
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern 67663, Germany.,German Research Center for Artificial Intelligence GmbH, Kaiserslautern 67663, Germany
| | - Ahtisham Fazeel
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern 67663, Germany.,German Research Center for Artificial Intelligence GmbH, Kaiserslautern 67663, Germany
| | - Andreas Dengel
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern 67663, Germany.,German Research Center for Artificial Intelligence GmbH, Kaiserslautern 67663, Germany
| | - Sheraz Ahmed
- German Research Center for Artificial Intelligence GmbH, Kaiserslautern 67663, Germany
| |
Collapse
|
41
|
DNA Modification Patterns Filtering and Analysis Using DNAModAnnot. Methods Mol Biol 2023; 2624:87-114. [PMID: 36723811 DOI: 10.1007/978-1-0716-2962-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mapping DNA modifications at the base resolution is now possible at the genome level thanks to advances in sequencing technologies. Long-read sequencing data can be used to identify modified base patterns. However, the downstream analysis of Pacific Biosciences (PacBio) or Oxford Nanopore Technologies (ONT) data requires the integration of genomic annotation and comprehensive filtering to prevent the accumulation of artifact signals. We present in this chapter, a linear workflow to fully analyze modified base patterns using the DNA Modification Annotation (DNAModAnnot) package. This workflow includes a thorough filtering based on sequencing quality and false discovery rate estimation and provides tools for a global analysis of DNA modifications. Here, we provide an application example of this workflow with PacBio data and guide the user by explaining expected outputs via a fully integrated Rmarkdown script. This protocol is presented with tips showing how to adapt the provided code for annotating epigenomes of any organism according to the user needs.
Collapse
|
42
|
Zeng W, Gautam A, Huson DH. MuLan-Methyl-multiple transformer-based language models for accurate DNA methylation prediction. Gigascience 2022; 12:giad054. [PMID: 37489753 PMCID: PMC10367125 DOI: 10.1093/gigascience/giad054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/09/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023] Open
Abstract
Transformer-based language models are successfully used to address massive text-related tasks. DNA methylation is an important epigenetic mechanism, and its analysis provides valuable insights into gene regulation and biomarker identification. Several deep learning-based methods have been proposed to identify DNA methylation, and each seeks to strike a balance between computational effort and accuracy. Here, we introduce MuLan-Methyl, a deep learning framework for predicting DNA methylation sites, which is based on 5 popular transformer-based language models. The framework identifies methylation sites for 3 different types of DNA methylation: N6-adenine, N4-cytosine, and 5-hydroxymethylcytosine. Each of the employed language models is adapted to the task using the "pretrain and fine-tune" paradigm. Pretraining is performed on a custom corpus of DNA fragments and taxonomy lineages using self-supervised learning. Fine-tuning aims at predicting the DNA methylation status of each type. The 5 models are used to collectively predict the DNA methylation status. We report excellent performance of MuLan-Methyl on a benchmark dataset. Moreover, we argue that the model captures characteristic differences between different species that are relevant for methylation. This work demonstrates that language models can be successfully adapted to applications in biological sequence analysis and that joint utilization of different language models improves model performance. Mulan-Methyl is open source, and we provide a web server that implements the approach.
Collapse
Affiliation(s)
- Wenhuan Zeng
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, 72076 Tübingen, Germany
| | - Anupam Gautam
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, 72076 Tübingen, Germany
- International Max Planck Research School “From Molecules to Organisms”, Max Planck Institute for Biology Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence: EXC 2124: Controlling Microbes to Fight Infection, University of Tübingen, 72076 Tübingen, Germany
| | - Daniel H Huson
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, 72076 Tübingen, Germany
- International Max Planck Research School “From Molecules to Organisms”, Max Planck Institute for Biology Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence: EXC 2124: Controlling Microbes to Fight Infection, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
43
|
Chen LQ, Zhang Z, Chen HX, Xi JF, Liu XH, Ma DZ, Zhong YH, Ng WH, Chen T, Mak DW, Chen Q, Chen YQ, Luo GZ. High-precision mapping reveals rare N 6-deoxyadenosine methylation in the mammalian genome. Cell Discov 2022; 8:138. [PMID: 36575183 PMCID: PMC9794812 DOI: 10.1038/s41421-022-00484-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 10/16/2022] [Indexed: 12/28/2022] Open
Abstract
N6-deoxyadenosine methylation (6mA) is the most widespread type of DNA modification in prokaryotes and is also abundantly distributed in some unicellular eukaryotes. However, 6mA levels are remarkably low in mammals. The lack of a precise and comprehensive mapping method has hindered more advanced investigations of 6mA. Here, we report a new method MM-seq (modification-induced mismatch sequencing) for genome-wide 6mA mapping based on a novel detection principle. We found that modified DNA bases are prone to form a local open region that allows capture by antibody, for example, via a DNA breathing or base-flipping mechanism. Specified endonuclease or exonuclease can recognize the antibody-stabilized mismatch-like structure and mark the exact modified sites for sequencing readout. Using this method, we examined the genomic positions of 6mA in bacteria (E. coli), green algae (C. reinhardtii), and mammalian cells (HEK239T, Huh7, and HeLa cells). In contrast to bacteria and green algae, human cells possess a very limited number of 6mA sites which are sporadically distributed across the genome of different cell types. After knocking out the RNA m6A methyltransferase METTL3 in mouse ES cells, 6mA becomes mostly diminished. Our results imply that rare 6mA in the mammalian genome is introduced by RNA m6A machinery via a non-targeted mechanism.
Collapse
Affiliation(s)
- Li-Qian Chen
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhang Zhang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hong-Xuan Chen
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian-Fei Xi
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xue-Hong Liu
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dong-Zhao Ma
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu-Hao Zhong
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wen Hui Ng
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tao Chen
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Daniel W Mak
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qi Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yao-Qing Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Guan-Zheng Luo
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
44
|
Varma SJ, Calvani E, Grüning NM, Messner CB, Grayson N, Capuano F, Mülleder M, Ralser M. Global analysis of cytosine and adenine DNA modifications across the tree of life. eLife 2022; 11:81002. [PMID: 35900202 PMCID: PMC9333990 DOI: 10.7554/elife.81002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 12/12/2022] Open
Abstract
Interpreting the function and metabolism of enzymatic DNA modifications requires both position-specific and global quantities. Sequencing-based techniques that deliver the former have become broadly accessible, but analytical methods for the global quantification of DNA modifications have thus far been applied mostly to individual problems. We established a mass spectrometric method for the sensitive and accurate quantification of multiple enzymatic DNA modifications. Then, we isolated DNA from 124 archean, bacterial, fungal, plant, and mammalian species, and several tissues and created a resource of global DNA modification quantities. Our dataset provides insights into the general nature of enzymatic DNA modifications, reveals unique biological cases, and provides complementary quantitative information to normalize and assess the accuracy of sequencing-based detection of DNA modifications. We report that only three of the studied DNA modifications, methylcytosine (5mdC), methyladenine (N6mdA) and hydroxymethylcytosine (5hmdC), were detected above a picomolar detection limit across species, and dominated in higher eukaryotes (5mdC), in bacteria (N6mdA), or the vertebrate central nervous systems (5hmdC). All three modifications were detected simultaneously in only one of the tested species, Raphanus sativus. In contrast, these modifications were either absent or detected only at trace quantities, across all yeasts and insect genomes studied. Further, we reveal interesting biological cases. For instance, in Allium cepa, Helianthus annuus, or Andropogon gerardi, more than 35% of cytosines were methylated. Additionally, next to the mammlian CNS, 5hmdC was also detected in plants like Lepidium sativum and was found on 8% of cytosines in the Garra barreimiae brain samples. Thus, identifying unexpected levels of DNA modifications in several wild species, our resource underscores the need to address biological diversity for studying DNA modifications.
Collapse
Affiliation(s)
| | - Enrica Calvani
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, United Kingdom.,Department of Biochemistry and Cambridge Systems Biology Center, University of Cambridge, Cambridge, United Kingdom
| | - Nana-Maria Grüning
- Department of Biochemistry, Charité Universitätsmedizin, Berlin, Germany
| | - Christoph B Messner
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, United Kingdom.,Department of Biochemistry and Cambridge Systems Biology Center, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas Grayson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Floriana Capuano
- Department of Biochemistry and Cambridge Systems Biology Center, University of Cambridge, Cambridge, United Kingdom
| | - Michael Mülleder
- Core Facility-High Throughput Mass Spectrometry, Charité Universitätsmedizin, Berlin, Germany
| | - Markus Ralser
- Department of Biochemistry, Charité Universitätsmedizin, Berlin, Germany.,The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, United Kingdom.,Department of Biochemistry and Cambridge Systems Biology Center, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
45
|
Moshareva MA, Lukyanov KA, Putlyaeva LV. Fluorescence imaging of epigenetic genome modifications. Biochem Biophys Res Commun 2022; 622:86-92. [PMID: 35843098 DOI: 10.1016/j.bbrc.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022]
Abstract
Epigenome contains a lot of information about cell state. Epigenetic analysis includes primarily sequence-based methods, which provide detailed data on distribution of modifications along the genome, but are poorly applicable for screenings. Specific fluorescence labeling and imaging of epigenetic modifications is an attractive complementary approach. It is currently based mainly on histone modifications study. We expect that inclusion of DNA modifications into imaging-based study would empower the method. In this review we discuss methods for fluorescence imaging of DNA modifications (mainly 5-methylcytosine). It opens an easy way to single cell analysis and high-throughput screening. Moreover, tracking epigenome changes in live cells becomes possible with genetically encoded probes.
Collapse
Affiliation(s)
- Maria A Moshareva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Konstantin A Lukyanov
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Lidia V Putlyaeva
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, Russia.
| |
Collapse
|
46
|
Sarkies P. Encyclopaedia of eukaryotic DNA methylation: from patterns to mechanisms and functions. Biochem Soc Trans 2022; 50:1179-1190. [PMID: 35521905 PMCID: PMC9246332 DOI: 10.1042/bst20210725] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/14/2022]
Abstract
DNA methylation is an epigenetic modification with a very long evolutionary history. However, DNA methylation evolves surprisingly rapidly across eukaryotes. The genome-wide distribution of methylation diversifies rapidly in different lineages, and DNA methylation is lost altogether surprisingly frequently. The growing availability of genomic and epigenomic sequencing across organisms highlights this diversity but also illuminates potential factors that could explain why both the DNA methylation machinery and its genome-wide distribution evolve so rapidly. Key to this are new discoveries about the fitness costs associated with DNA methylation, and new theories about how the fundamental biochemical mechanisms of DNA methylation introduction and maintenance could explain how new genome-wide patterns of methylation evolve.
Collapse
Affiliation(s)
- Peter Sarkies
- Department of Biochemistry, University of Oxford, Oxford, U.K
- MRC London Institute of Molecular Biology, London, U.K
- Institute of Clinical Sciences, Imperial College London, London, U.K
| |
Collapse
|
47
|
Li H, Zhang N, Wang Y, Xia S, Zhu Y, Xing C, Tian X, Du Y. DNA N6-Methyladenine Modification in Eukaryotic Genome. Front Genet 2022; 13:914404. [PMID: 35812743 PMCID: PMC9263368 DOI: 10.3389/fgene.2022.914404] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022] Open
Abstract
DNA methylation is treated as an important epigenetic mark in various biological activities. In the past, a large number of articles focused on 5 mC while lacking attention to N6-methyladenine (6 mA). The presence of 6 mA modification was previously discovered only in prokaryotes. Recently, with the development of detection technologies, 6 mA has been found in several eukaryotes, including protozoans, metazoans, plants, and fungi. The importance of 6 mA in prokaryotes and single-celled eukaryotes has been widely accepted. However, due to the incredibly low density of 6 mA and restrictions on detection technologies, the prevalence of 6 mA and its role in biological processes in eukaryotic organisms are highly debated. In this review, we first summarize the advantages and disadvantages of 6 mA detection methods. Then, we conclude existing reports on the prevalence of 6 mA in eukaryotic organisms. Next, we highlight possible methyltransferases, demethylases, and the recognition proteins of 6 mA. In addition, we summarize the functions of 6 mA in eukaryotes. Last but not least, we summarize our point of view and put forward the problems that need further research.
Collapse
Affiliation(s)
- Hao Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- First School of Clinical Medicine, Anhui Medical University, Hefei, China
- First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ning Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- First School of Clinical Medicine, Anhui Medical University, Hefei, China
- First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuechen Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Second School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Siyuan Xia
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Second School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Yating Zhu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Chen Xing
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xuefeng Tian
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- First School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Yinan Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Yinan Du,
| |
Collapse
|
48
|
Jiménez-Ramírez IA, Pijeira-Fernández G, Moreno-Cálix DM, De-la-Peña C. Same modification, different location: the mythical role of N 6-adenine methylation in plant genomes. PLANTA 2022; 256:9. [PMID: 35696004 DOI: 10.1007/s00425-022-03926-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/23/2022] [Indexed: 06/15/2023]
Abstract
The present review summarizes recent advances in the understanding of 6mA in DNA as an emergent epigenetic mark with distinctive characteristics, discusses its importance in plant genomes, and highlights its chemical nature and functions. Adenine methylation is an epigenetic modification present in DNA (6mA) and RNA (m6A) that has a regulatory function in many cellular processes. This modification occurs through a reversible reaction that covalently binds a methyl group, usually at the N6 position of the purine ring. This modification carries biophysical properties that affect the stability of nucleic acids as well as their binding affinity with other molecules. DNA 6mA has been related to genome stability, gene expression, DNA replication, and repair mechanisms. Recent advances have shown that 6mA in plant genomes is related to development and stress response. In this review, we present recent advances in the understanding of 6mA in DNA as an emergent epigenetic mark with distinctive characteristics. We discuss the key elements of this modification, focusing mainly on its importance in plant genomes. Furthermore, we highlight its chemical nature and the regulatory effects that it exerts on gene expression and plant development. Finally, we emphasize the functions of 6mA in photosynthesis, stress, and flowering.
Collapse
Affiliation(s)
- Irma A Jiménez-Ramírez
- Unidad de Bioquímica y Biología Molecular de Plantas, Centro de Investigación Científica de Yucatán, Calle 43 No. 130 x 32 y 34. Col. Chuburná de Hidalgo, 97205, Mérida, Yucatán, Mexico
| | - Gema Pijeira-Fernández
- Unidad de Bioquímica y Biología Molecular de Plantas, Centro de Investigación Científica de Yucatán, Calle 43 No. 130 x 32 y 34. Col. Chuburná de Hidalgo, 97205, Mérida, Yucatán, Mexico
| | - Delia M Moreno-Cálix
- Unidad de Bioquímica y Biología Molecular de Plantas, Centro de Investigación Científica de Yucatán, Calle 43 No. 130 x 32 y 34. Col. Chuburná de Hidalgo, 97205, Mérida, Yucatán, Mexico
| | - Clelia De-la-Peña
- Centro de Investigación Científica de Yucatán, Unidad de Biotecnología, Calle 43 No. 130 x 32 y 34. Col. Chuburná de Hidalgo, 97205, Mérida, Yucatán, Mexico.
| |
Collapse
|
49
|
A fungal dioxygenase CcTet serves as a eukaryotic 6mA demethylase on duplex DNA. Nat Chem Biol 2022; 18:733-741. [PMID: 35654845 DOI: 10.1038/s41589-022-01041-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 03/31/2022] [Indexed: 12/24/2022]
Abstract
N6-methyladenosine (6mA) is a DNA modification that has recently been found to play regulatory roles during mammalian early embryo development and mitochondrial transcription. We found that a dioxygenase CcTet from the fungus Coprinopsis cinerea is also a dsDNA 6mA demethylase. It oxidizes 6mA to the intermediate N6-hydroxymethyladenosine (6hmA) with robust activity of 6mA-containing duplex DNA (dsDNA) as well as isolated genomics DNA. Structural characterization revealed that CcTet utilizes three flexible loop regions and two key residues-D337 and G331-in the active pocket to preferentially recognize substrates on dsDNA. A CcTet D337F mutant protein retained the catalytic activity on 6mA but lost activity on 5-methylcytosine. Our findings uncovered a 6mA demethylase that works on dsDNA, suggesting potential 6mA demethylation in fungi and elucidating 6mA recognition and the catalytic mechanism of CcTet. The CcTet D337F mutant protein also provides a chemical biology tool for future functional manipulation of DNA 6mA in vivo.
Collapse
|
50
|
Altemose N, Maslan A, Smith OK, Sundararajan K, Brown RR, Mishra R, Detweiler AM, Neff N, Miga KH, Straight AF, Streets A. DiMeLo-seq: a long-read, single-molecule method for mapping protein-DNA interactions genome wide. Nat Methods 2022; 19:711-723. [PMID: 35396487 PMCID: PMC9189060 DOI: 10.1038/s41592-022-01475-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/24/2022] [Indexed: 12/13/2022]
Abstract
Studies of genome regulation routinely use high-throughput DNA sequencing approaches to determine where specific proteins interact with DNA, and they rely on DNA amplification and short-read sequencing, limiting their quantitative application in complex genomic regions. To address these limitations, we developed directed methylation with long-read sequencing (DiMeLo-seq), which uses antibody-tethered enzymes to methylate DNA near a target protein's binding sites in situ. These exogenous methylation marks are then detected simultaneously with endogenous CpG methylation on unamplified DNA using long-read, single-molecule sequencing technologies. We optimized and benchmarked DiMeLo-seq by mapping chromatin-binding proteins and histone modifications across the human genome. Furthermore, we identified where centromere protein A localizes within highly repetitive regions that were unmappable with short sequencing reads, and we estimated the density of centromere protein A molecules along single chromatin fibers. DiMeLo-seq is a versatile method that provides multimodal, genome-wide information for investigating protein-DNA interactions.
Collapse
Affiliation(s)
- Nicolas Altemose
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Annie Maslan
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Owen K Smith
- Department of Biochemistry, Stanford University, Stanford, CA, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | | | - Rachel R Brown
- Department of Biochemistry, Stanford University, Stanford, CA, USA
| | - Reet Mishra
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | | | - Norma Neff
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Karen H Miga
- Department of Molecular & Cell Biology, University of California, Santa Cruz, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Aaron F Straight
- Department of Biochemistry, Stanford University, Stanford, CA, USA.
| | - Aaron Streets
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA.
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, USA.
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|