RNA N6-methyladenosine modulates endothelial atherogenic responses to disturbed flow in mice

  1. Bochuan Li
  2. Ting Zhang
  3. Mengxia Liu
  4. Zhen Cui
  5. Yanhong Zhang
  6. Mingming Liu
  7. Yanan Liu
  8. Yongqiao Sun
  9. Mengqi Li
  10. Yikui Tian
  11. Ying Yang  Is a corresponding author
  12. Hongfeng Jiang  Is a corresponding author
  13. Degang Liang  Is a corresponding author
  1. Tianjin Medical University, China
  2. Chinese Academy of Sciences, China
  3. Capital Medical University, China

Abstract

Atherosclerosis preferentially occurs in atheroprone vasculature where human umbilical vein endothelial cells (HUVECs) are exposed to disturbed flow. Disturbed flow is associated with vascular inflammation and focal distribution. Recent studies have revealed the involvement of epigenetic regulation in atherosclerosis progression. N6-methyladenosine (m6A) is the most prevalent internal modification of eukaryotic mRNA, but its function in endothelial atherogenic progression remains unclear. Here, we show that m6A mediates the EGFR signaling pathway during EC activation to regulate the atherosclerotic process. Oscillatory stress (OS) reduced the expression of METTL3, the primary m6A methyltransferase. Through m6A sequencing and functional studies, we determined that m6A mediates the mRNA decay of the vascular pathophysiology gene EGFR which leads to EC dysfunction. m6A modification of the EGFR 3'UTR accelerated its mRNA degradation. Double mutation of the EGFR 3'UTR abolished METTL3-induced luciferase activity. Adenovirus-mediated METTL3 overexpression significantly reduced EGFR activation and endothelial dysfunction in the presence of OS. Furthermore, TSP-1, an EGFR ligand, was specifically expressed in atheroprone regions without being affected by METTL3. Inhibition of the TSP-1/EGFR axis by using shRNA and AG1478 significantly ameliorated atherogenesis. Overall, our study revealed that METTL3 alleviates endothelial atherogenic progression through m6A-dependent stabilization of EGFR mRNA, highlighting the important role of RNA transcriptomics in atherosclerosis regulation.

Data availability

RNA-seq and MeRIP-seq data generated in this study have been deposited to the Genome Sequence Archive in BIG Data Center under accession number PRJCA004746.

The following data sets were generated

Article and author information

Author details

  1. Bochuan Li

    Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
    Competing interests
    The authors declare that no competing interests exist.
  2. Ting Zhang

    CAS Key Laboratory of Genomic and Precision Medicine, Chinese Academy of Sciences, Beijing, China
    Competing interests
    The authors declare that no competing interests exist.
  3. Mengxia Liu

    CAS Key Laboratory of Genomic and Precision Medicine, Chinese Academy of Sciences, Beijing, China
    Competing interests
    The authors declare that no competing interests exist.
  4. Zhen Cui

    Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
    Competing interests
    The authors declare that no competing interests exist.
  5. Yanhong Zhang

    Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
    Competing interests
    The authors declare that no competing interests exist.
  6. Mingming Liu

    Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
    Competing interests
    The authors declare that no competing interests exist.
  7. Yanan Liu

    Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
    Competing interests
    The authors declare that no competing interests exist.
  8. Yongqiao Sun

    CAS Key Laboratory of Genomic and Precision Medicine, Chinese Academy of Sciences, Beijing, China
    Competing interests
    The authors declare that no competing interests exist.
  9. Mengqi Li

    Tianjin Medical University General Hospital Cardiovascular Department, Tianjin Medical University, Tianjin, China
    Competing interests
    The authors declare that no competing interests exist.
  10. Yikui Tian

    Tianjin Medical University General Hospital Cardiovascular Department, Tianjin Medical University, Tianjin, China
    Competing interests
    The authors declare that no competing interests exist.
  11. Ying Yang

    CAS Key Laboratory of Genomic and Precision Medicine, Chinese Academy of Sciences, Beijing, China
    For correspondence
    yingyang@big.ac.cn
    Competing interests
    The authors declare that no competing interests exist.
  12. Hongfeng Jiang

    Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Tianjin, China
    For correspondence
    jhf@pku.edu.cn
    Competing interests
    The authors declare that no competing interests exist.
  13. Degang Liang

    Tianjin Medical University General Hospital Cardiovascular Department, Tianjin Medical University, Tianjin, China
    For correspondence
    15922230066@163.com
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-2618-6651

Funding

National Natural Science Foundation of China (81900396)

  • Bochuan Li

National Natural Science Foundation of China (82000477)

  • Mengqi Li

National Natural Science Foundation of China (81970392)

  • Hongfeng Jiang

Postdoctoral Research Foundation of China (2019M661041)

  • Bochuan Li

Postdoctoral Research Foundation of China (BX20190235)

  • Bochuan Li

China Association for Science and Technology (Excellent Sino-foreign Youth Exchange Program)

  • Bochuan Li

National Natural Science Foundation of China (91940304)

  • Ying Yang

Chinese Academy of Sciences (2018133)

  • Ying Yang

National Key Research and Development Program of China (2018YFA0801200)

  • Ying Yang

Beijing Nova Program (Z201100006820104)

  • Ying Yang

National Natural Science Foundation of China (81870207)

  • Yikui Tian

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Reviewing Editor

  1. Edward A Fisher, New York University Grossman School of Medicine, United States

Ethics

Animal experimentation: The investigation conformed to the Guide for the Care and Use of Laboratory Animals by the US National Institutes of Health (NIH 17 Publication No. 85-23, revised in 2011). All study protocols and the use of animals were approved by the Institutional Animal Care and Use Committee of Tianjin Medical University.

Version history

  1. Received: April 29, 2021
  2. Accepted: January 7, 2022
  3. Accepted Manuscript published: January 10, 2022 (version 1)
  4. Version of Record published: January 27, 2022 (version 2)

Copyright

© 2022, Li et al.

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 974
    views
  • 168
    downloads
  • 14
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Bochuan Li
  2. Ting Zhang
  3. Mengxia Liu
  4. Zhen Cui
  5. Yanhong Zhang
  6. Mingming Liu
  7. Yanan Liu
  8. Yongqiao Sun
  9. Mengqi Li
  10. Yikui Tian
  11. Ying Yang
  12. Hongfeng Jiang
  13. Degang Liang
(2022)
RNA N6-methyladenosine modulates endothelial atherogenic responses to disturbed flow in mice
eLife 11:e69906.
https://doi.org/10.7554/eLife.69906

Share this article

https://doi.org/10.7554/eLife.69906

Further reading

    1. Cell Biology
    2. Developmental Biology
    Filip Knop, Apolena Zounarova ... Marie Macůrková
    Research Article

    During Caenorhabditis elegans development, multiple cells migrate long distances or extend processes to reach their final position and/or attain proper shape. The Wnt signalling pathway stands out as one of the major coordinators of cell migration or cell outgrowth along the anterior-posterior body axis. The outcome of Wnt signalling is fine-tuned by various mechanisms including endocytosis. In this study, we show that SEL-5, the C. elegans orthologue of mammalian AP2-associated kinase AAK1, acts together with the retromer complex as a positive regulator of EGL-20/Wnt signalling during the migration of QL neuroblast daughter cells. At the same time, SEL-5 in cooperation with the retromer complex is also required during excretory canal cell outgrowth. Importantly, SEL-5 kinase activity is not required for its role in neuronal migration or excretory cell outgrowth, and neither of these processes is dependent on DPY-23/AP2M1 phosphorylation. We further establish that the Wnt proteins CWN-1 and CWN-2 together with the Frizzled receptor CFZ-2 positively regulate excretory cell outgrowth, while LIN-44/Wnt and LIN-17/Frizzled together generate a stop signal inhibiting its extension.

    1. Biochemistry and Chemical Biology
    2. Cell Biology
    Ya-Juan Wang, Xiao-Jing Di ... Ting-Wei Mu
    Research Article Updated

    Protein homeostasis (proteostasis) deficiency is an important contributing factor to neurological and metabolic diseases. However, how the proteostasis network orchestrates the folding and assembly of multi-subunit membrane proteins is poorly understood. Previous proteomics studies identified Hsp47 (Gene: SERPINH1), a heat shock protein in the endoplasmic reticulum lumen, as the most enriched interacting chaperone for gamma-aminobutyric acid type A (GABAA) receptors. Here, we show that Hsp47 enhances the functional surface expression of GABAA receptors in rat neurons and human HEK293T cells. Furthermore, molecular mechanism study demonstrates that Hsp47 acts after BiP (Gene: HSPA5) and preferentially binds the folded conformation of GABAA receptors without inducing the unfolded protein response in HEK293T cells. Therefore, Hsp47 promotes the subunit-subunit interaction, the receptor assembly process, and the anterograde trafficking of GABAA receptors. Overexpressing Hsp47 is sufficient to correct the surface expression and function of epilepsy-associated GABAA receptor variants in HEK293T cells. Hsp47 also promotes the surface trafficking of other Cys-loop receptors, including nicotinic acetylcholine receptors and serotonin type 3 receptors in HEK293T cells. Therefore, in addition to its known function as a collagen chaperone, this work establishes that Hsp47 plays a critical and general role in the maturation of multi-subunit Cys-loop neuroreceptors.