ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Figure 1Loading Img

A High-Throughput Quantitative Approach Reveals More Small RNA Modifications in Mouse Liver and Their Correlation with Diabetes

View Author Information
Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
*Phone: +86 21 5492 0903. Fax: +86 21 5492 0291. E-mail: [email protected]
Cite this: Anal. Chem. 2013, 85, 24, 12173–12181
Publication Date (Web):November 21, 2013
https://doi.org/10.1021/ac4036026
Copyright © 2013 American Chemical Society

    Article Views

    1821

    Altmetric

    -

    Citations

    44
    LEARN ABOUT THESE METRICS
    Other access options
    Supporting Info (1)»

    Abstract

    Abstract Image

    Studies of RNA modification are usually focused on tRNA. However the modification of other small RNAs, including 5.8S rRNA, 5S rRNA, and small RNA sized at 10–60 nt, is still largely unknown. In this study, we established an efficient method based on liquid chromatography–tandem mass spectrometry (LC–MS/MS) to simultaneously identify and quantify more than 40 different types of nucleosides in small RNAs. With this method, we revealed 23 modified nucleosides of tRNA from mouse liver, and 6 of them were observed for the first time in eukaryotic tRNA. Moreover, 5 and 4 modified nucleosides were detected for the first time in eukaryotic 5.8S and 5S rRNA, respectively, and 22 modified nucleosides were identified in the small RNAs sized at 30–60 or 10–30 nt. Interestingly, two groups of 5S rRNA peaks were observed when analyzed by HPLC, and the abundance of modified nucleosides is significantly different between the two groups of peaks. Further studies show that multiple modifications in small RNA from diabetic mouse liver are significantly increased or decreased. Taken together, our data revealed more modified nucleosides in various small RNAs and showed the correlation of small RNA modifications with diabetes. These results provide new insights to the role of modifications of small RNAs in their stability, biological functions, and correlation with diseases.

    Read this article

    To access this article, please review the available access options below.

    Get instant access

    Purchase Access

    Read this article for 48 hours. Check out below using your ACS ID or as a guest.

    Recommended

    Access through Your Institution

    You may have access to this article through your institution.

    Your institution does not have access to this content. You can change your affiliated institution below.

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    Additional information as noted in text. This material is available free of charge via the Internet at http://pubs.acs.org.

    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

    Cited By

    This article is cited by 44 publications.

    1. Clarisse Gosset-Erard, Mévie Didierjean, Jérome Pansanel, Antony Lechner, Philippe Wolff, Lauriane Kuhn, Frédéric Aubriet, Emmanuelle Leize-Wagner, Patrick Chaimbault, Yannis-Nicolas François. Nucleos’ID: A New Search Engine Enabling the Untargeted Identification of RNA Post-transcriptional Modifications from Tandem Mass Spectrometry Analyses of Nucleosides. Analytical Chemistry 2023, 95 (2) , 1608-1617. https://doi.org/10.1021/acs.analchem.2c04722
    2. Yixuan Xie, Kevin A. Janssen, Alessandro Scacchetti, Elizabeth G. Porter, Zongtao Lin, Roberto Bonasio, Benjamin A. Garcia. Permethylation of Ribonucleosides Provides Enhanced Mass Spectrometry Quantification of Post-Transcriptional RNA Modifications. Analytical Chemistry 2022, 94 (20) , 7246-7254. https://doi.org/10.1021/acs.analchem.2c00471
    3. Wanqiao Bai, Aiping Cui, Meizhou Liu, Xuezhi Qiao, Yan Li, Tie Wang. Signal-Off Electrogenerated Chemiluminescence Biosensing Platform Based on the Quenching Effect between Ferrocene and Ru(bpy)32+-Functionalized Metal–Organic Frameworks for the Detection of Methylated RNA. Analytical Chemistry 2019, 91 (18) , 11840-11847. https://doi.org/10.1021/acs.analchem.9b02569
    4. Lijuan Fu, Nicholas J. Amato, Pengcheng Wang, Sara J. McGowan, Laura J. Niedernhofer, and Yinsheng Wang . Simultaneous Quantification of Methylated Cytidine and Adenosine in Cellular and Tissue RNA by Nano-Flow Liquid Chromatography–Tandem Mass Spectrometry Coupled with the Stable Isotope-Dilution Method. Analytical Chemistry 2015, 87 (15) , 7653-7659. https://doi.org/10.1021/acs.analchem.5b00951
    5. Karine Dubuc, Mathilde Marchais, Isabelle Gilbert, Alexandre Bastien, Karen E. Nenonene, Edward W. Khandjian, Robert S. Viger, Géraldine Delbes, Claude Robert. Epitranscriptome marks detection and localization of RNA modifying proteins in mammalian ovarian follicles. Journal of Ovarian Research 2023, 16 (1) https://doi.org/10.1186/s13048-023-01172-8
    6. Qunli Xiong, Yaguang Zhang. Small RNA modifications: regulatory molecules and potential applications. Journal of Hematology & Oncology 2023, 16 (1) https://doi.org/10.1186/s13045-023-01466-w
    7. Hejin Lai, Ning Feng, Qiwei Zhai. Discovery of the major 15–30 nt mammalian small RNAs, their biogenesis and function. Nature Communications 2023, 14 (1) https://doi.org/10.1038/s41467-023-41554-6
    8. Gefei Huang, Feng Zhang, Dongying Xie, Yiming Ma, Pengxi Wang, Guodong Cao, Leijian Chen, Siyi Lin, Zhongying Zhao, Zongwei Cai. High-throughput profiling of RNA modifications by ultra-performance liquid chromatography coupled to complementary mass spectrometry: Methods, quality control, and applications. Talanta 2023, 263 , 124697. https://doi.org/10.1016/j.talanta.2023.124697
    9. Gang Li, Nawaraj Dulal, Ziwen Gong, Richard A. Wilson. Unconventional secretion of Magnaporthe oryzae effectors in rice cells is regulated by tRNA modification and codon usage control. Nature Microbiology 2023, 8 (9) , 1706-1716. https://doi.org/10.1038/s41564-023-01443-6
    10. Yan Li, Yumeng Sun, Weiguo Yang, Lan Yang, Mingli Su, Lichao Fang, Junsong Zheng, Ruo Yuan, Wenbin Liang. A novel photoelectrochemical strategy for sequence-spot bispecific analysis of N6-methyladenosine modification based on proximity ligation-triggered cascade amplification. Analytica Chimica Acta 2023, 1265 , 341287. https://doi.org/10.1016/j.aca.2023.341287
    11. Haiping Li, Zhikun Zhang, Lu Gan, Dianfa Fan, Xinjun Sun, Zhangbo Qian, Xiyu Liu, Yong Huang. Signal Amplification-Based Biosensors and Application in RNA Tumor Markers. Sensors 2023, 23 (9) , 4237. https://doi.org/10.3390/s23094237
    12. Taylor A. Dodson, Stephan Nieuwoudt, Chase N. Morse, Valinteshley Pierre, Chao Liu, Samuel E. Senyo, Erin G. Prestwich. Ribonucleosides from tRNA in hyperglycemic mammalian cells and diabetic murine cardiac models. Life Sciences 2023, 318 , 121462. https://doi.org/10.1016/j.lfs.2023.121462
    13. Serafima Dubnov, Hermona Soreq. Transfer RNA Fragments, from Structure to Function. 2023, 1-19. https://doi.org/10.1007/978-3-031-36390-0_1
    14. Lu Chen, Wen-Jing Wang, Qiang Liu, Yu-Ke Wu, Yun-Wen Wu, Yu Jiang, Xiu-Quan Liao, Fei Huang, Yang Li, Li Shen, Chao Yu, Song-Ying Zhang, Li-Ying Yan, Jie Qiao, Qian-Qian Sha, Heng-Yu Fan. NAT10-mediated N 4-acetylcytidine modification is required for meiosis entry and progression in male germ cells. Nucleic Acids Research 2022, 50 (19) , 10896-10913. https://doi.org/10.1093/nar/gkac594
    15. Dandan Xu, Deqian Qiao, Yunlong Lei, Chundong Zhang, Youquan Bu, Ying Zhang. Transfer RNA-derived small RNAs (tsRNAs): Versatile regulators in cancer. Cancer Letters 2022, 546 , 215842. https://doi.org/10.1016/j.canlet.2022.215842
    16. Clarisse Gosset-Erard, Antony Lechner, Philippe Wolff, Frédéric Aubriet, Emmanuelle Leize-Wagner, Patrick Chaimbault, Yannis-Nicolas François. Optimization of nucleotides dephosphorylation for RNA structural characterization by tandem mass spectrometry hyphenated with separation methods. Journal of Chromatography B 2022, 1208 , 123396. https://doi.org/10.1016/j.jchromb.2022.123396
    17. Zhihang Chen, Wanjie Zhu, Shenghua Zhu, Kaiyu Sun, Junbin Liao, Haining Liu, Zihao Dai, Hui Han, Xuxin Ren, Qingxia Yang, Siyi Zheng, Baogang Peng, Sui Peng, Ming Kuang, Shuibin Lin. METTL1 promotes hepatocarcinogenesis via m 7 G tRNA modification‐dependent translation control. Clinical and Translational Medicine 2021, 11 (12) https://doi.org/10.1002/ctm2.661
    18. Pavlina Gregorova, Nina H. Sipari, L. Peter Sarin. Broad-range RNA modification analysis of complex biological samples using rapid C18-UPLC-MS. RNA Biology 2021, 18 (10) , 1382-1389. https://doi.org/10.1080/15476286.2020.1853385
    19. Jing Li, Wen-Yu Zhu, Wen-Qing Yang, Cai-Tao Li, Ru-Juan Liu. The occurrence order and cross-talk of different tRNA modifications. Science China Life Sciences 2021, 64 (9) , 1423-1436. https://doi.org/10.1007/s11427-020-1906-4
    20. Hongmei Yang, Yafen Wang, Jing Tang, Fang Wang, Zilin Chen. End-labeling-based electrochemical strategy for detection of adenine methylation in nucleic acid by differential pulse voltammetry. Microchimica Acta 2021, 188 (8) https://doi.org/10.1007/s00604-021-04898-8
    21. Priti Thakur, Manasses Jora, Ruoxia Zhao, Gwenn Parungao, Scott Abernathy, Patrick A. Limbach, Balasubrahmanyam Addepalli. Mass Spectrometry-Based Methods for Characterization of Hypomodifications in Transfer RNA. 2021, 555-592. https://doi.org/10.1007/978-3-030-71612-7_21
    22. Hailang Wang, Chao Xu, Youbing Zhang, Xu Yan, Xiaohuan Jin, Xiaoqing Yao, Peng Chen, Bo Zheng, . PtKTI12 genes influence wobble uridine modifications and drought stress tolerance in hybrid poplar. Tree Physiology 2020, 40 (12) , 1778-1791. https://doi.org/10.1093/treephys/tpaa088
    23. Hongwei Liang, Zichen Jiao, Weiwei Rong, Shuang Qu, Zhicong Liao, Xinlei Sun, Yao Wei, Quan Zhao, Jun Wang, Yuan Liu, Xi Chen, Tao Wang, Chen-Yu Zhang, Ke Zen. 3′-Terminal 2′-O-methylation of lung cancer miR-21-5p enhances its stability and association with Argonaute 2. Nucleic Acids Research 2020, 18 https://doi.org/10.1093/nar/gkaa504
    24. Jun Tang, Pengfei Jia, Peiyong Xin, Jinfang Chu, Dong-Qiao Shi, Wei-Cai Yang, . The Arabidopsis TRM61/TRM6 complex is a bona fide tRNA N1-methyladenosine methyltransferase. Journal of Experimental Botany 2020, 71 (10) , 3024-3036. https://doi.org/10.1093/jxb/eraa100
    25. Qihan He, Lin Yang, Kaiping Gao, Peikun Ding, Qianqian Chen, Juan Xiong, Wenhan Yang, Yi Song, Liang Wang, Yejun Wang, Lijuan Ling, Weiming Wu, Jisong Yan, Peng Zou, Yuchen Chen, Rihong Zhai. FTSJ1 regulates tRNA 2ʹ-O-methyladenosine modification and suppresses the malignancy of NSCLC via inhibiting DRAM1 expression. Cell Death & Disease 2020, 11 (5) https://doi.org/10.1038/s41419-020-2525-x
    26. Xinying Ou, Qinli Pu, Shangchun Sheng, Tao Dai, Dan Gou, Wen Yu, Tingyan Yang, Ling Dai, Yujun Yang, Guoming Xie. Electrochemical competitive immunodetection of messenger RNA modified with N6-methyladenosine by using DNA-modified mesoporous PtCo nanospheres. Microchimica Acta 2020, 187 (1) https://doi.org/10.1007/s00604-019-4010-8
    27. Lei Su, Mary Elizabeth Patti. Paternal Nongenetic Intergenerational Transmission of Metabolic Disease Risk. Current Diabetes Reports 2019, 19 (7) https://doi.org/10.1007/s11892-019-1163-0
    28. Zheng Zhou, Bao Sun, Shiqiong Huang, Wenrui Jia, Dongsheng Yu. The tRNA-associated dysregulation in diabetes mellitus. Metabolism 2019, 94 , 9-17. https://doi.org/10.1016/j.metabol.2019.01.017
    29. Chubo Qi, Hanpeng Jiang, Jun Xiong, Bifeng Yuan, Yuqi Feng. On-line trapping/capillary hydrophilic-interaction liquid chromatography/mass spectrometry for sensitive determination of RNA modifications from human blood. Chinese Chemical Letters 2019, 30 (3) , 553-557. https://doi.org/10.1016/j.cclet.2018.11.029
    30. Xiaohuan Jin, Zhengyi Lv, Junbao Gao, Rui Zhang, Ting Zheng, Ping Yin, Dongqin Li, Liangcai Peng, Xintao Cao, Yan Qin, Staffan Persson, Bo Zheng, Peng Chen. AtTrm5a catalyses 1-methylguanosine and 1-methylinosine formation on tRNAs and is important for vegetative and reproductive growth in Arabidopsis thaliana. Nucleic Acids Research 2019, 47 (2) , 883-898. https://doi.org/10.1093/nar/gky1205
    31. Zhurui Li, Jing Shi, Lu Yu, Xiaozhen Zhao, Longlu Ran, Deyu Hu, Baoan Song. N 6 -methyl-adenosine level in Nicotiana tabacum is associated with tobacco mosaic virus. Virology Journal 2018, 15 (1) https://doi.org/10.1186/s12985-018-0997-4
    32. Tao Dai, Qinli Pu, Yongcan Guo, Chen Zuo, Shulian Bai, Yujun Yang, Dan Yin, Yi Li, Shangchun Sheng, Yiyi Tao, Jie Fang, Wen Yu, Guoming Xie. Analogous modified DNA probe and immune competition method-based electrochemical biosensor for RNA modification. Biosensors and Bioelectronics 2018, 114 , 72-77. https://doi.org/10.1016/j.bios.2018.05.018
    33. Dana Moravcová, Josef Planeta, Alistair W.T. King, Susanne K. Wiedmer. Immobilization of a phosphonium ionic liquid on a silica monolith for hydrophilic interaction chromatography. Journal of Chromatography A 2018, 1552 , 53-59. https://doi.org/10.1016/j.chroma.2018.04.012
    34. Ting Liu, Cheng-Jie Ma, Bi-Feng Yuan, Yu-Qi Feng. Modificaomics: deciphering the functions of biomolecule modifications. Science China Chemistry 2018, 61 (4) , 381-392. https://doi.org/10.1007/s11426-017-9186-y
    35. Nicky Jonkhout, Julia Tran, Martin A. Smith, Nicole Schonrock, John S. Mattick, Eva Maria Novoa. The RNA modification landscape in human disease. RNA 2017, 23 (12) , 1754-1769. https://doi.org/10.1261/rna.063503.117
    36. Heidelinde Glasner, Christian Riml, Ronald Micura, Kathrin Breuker. Label-free, direct localization and relative quantitation of the RNA nucleobase methylations m6A, m5C, m3U, and m5U by top-down mass spectrometry. Nucleic Acids Research 2017, 45 (13) , 8014-8025. https://doi.org/10.1093/nar/gkx470
    37. Youmei Wang, Dongqin Li, Junbao Gao, Xukai Li, Rui Zhang, Xiaohuan Jin, Zhen Hu, Bo Zheng, Staffan Persson, Peng Chen. The 2′-O-methyladenosine nucleoside modification gene OsTRM13 positively regulates salt stress tolerance in rice. Journal of Experimental Botany 2017, 68 (7) , 1479-1491. https://doi.org/10.1093/jxb/erx061
    38. Xudong Zhang, Aaron E. Cozen, Ying Liu, Qi Chen, Todd M. Lowe. Small RNA Modifications: Integral to Function and Disease. Trends in Molecular Medicine 2016, 22 (12) , 1025-1034. https://doi.org/10.1016/j.molmed.2016.10.009
    39. Qi Chen, Wei Yan, Enkui Duan. Epigenetic inheritance of acquired traits through sperm RNAs and sperm RNA modifications. Nature Reviews Genetics 2016, 17 (12) , 733-743. https://doi.org/10.1038/nrg.2016.106
    40. Yong Zhang, Josue Regalado, Wei Yan. Fat dad, fat kids: sperm small RNAs in control!. Science Bulletin 2016, 61 (6) , 428-429. https://doi.org/10.1007/s11434-016-1031-0
    41. Qi Chen, Menghong Yan, Zhonghong Cao, Xin Li, Yunfang Zhang, Junchao Shi, Gui-hai Feng, Hongying Peng, Xudong Zhang, Ying Zhang, Jingjing Qian, Enkui Duan, Qiwei Zhai, Qi Zhou. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science 2016, 351 (6271) , 397-400. https://doi.org/10.1126/science.aad7977
    42. John S. Satterlee, Maria Basanta-Sanchez, Sandra Blanco, Jin Billy Li, Kate Meyer, Jonathan Pollock, Ghazaleh Sadri-Vakili, Agnieszka Rybak-Wolf. Novel RNA Modifications in the Nervous System: Form and Function. The Journal of Neuroscience 2014, 34 (46) , 15170-15177. https://doi.org/10.1523/JNEUROSCI.3236-14.2014
    43. Stefanie Kellner, Antonia Ochel, Kathrin Thüring, Felix Spenkuch, Jennifer Neumann, Sunny Sharma, Karl-Dieter Entian, Dirk Schneider, Mark Helm. Absolute and relative quantification of RNA modifications via biosynthetic isotopomers. Nucleic Acids Research 2014, 42 (18) , e142-e142. https://doi.org/10.1093/nar/gku733
    44. David Gillis, Aiswarya Krishnamohan, Barak Yaacov, Avraham Shaag, Jane E Jackman, Orly Elpeleg. TRMT10A dysfunction is associated with abnormalities in glucose homeostasis, short stature and microcephaly. Journal of Medical Genetics 2014, 51 (9) , 581-586. https://doi.org/10.1136/jmedgenet-2014-102282

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    You’ve supercharged your research process with ACS and Mendeley!

    STEP 1:
    Click to create an ACS ID

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    MENDELEY PAIRING EXPIRED
    Your Mendeley pairing has expired. Please reconnect