Inhibition of DNMT1 methyltransferase activity via glucose-regulated O-GlcNAcylation alters the epigenome

Abstract

The DNA methyltransferase activity of DNMT1 is vital for genomic maintenance of DNA methylation. We report here that DNMT1 function is regulated by O-GlcNAcylation, a protein modification that is sensitive to glucose levels, and that elevated O-GlcNAcylation of DNMT1 from high glucose environment leads to alterations to the epigenome. Using mass spectrometry and complementary alanine mutation experiments, we identified S878 as the major residue that is O-GlcNAcylated on human DNMT1. Functional studies in human and mouse cells further revealed that O-GlcNAcylation of DNMT1-S878 results in an inhibition of methyltransferase activity, resulting in a general loss of DNA methylation that preferentially occurs at partially methylated domains (PMDs). This loss of methylation corresponds with an increase in DNA damage and apoptosis. These results establish O-GlcNAcylation of DNMT1 as a mechanism through which the epigenome is regulated by glucose metabolism and implicates a role for glycosylation of DNMT1 in metabolic diseases characterized by hyperglycemia.

Data availability

PromethION sequencing data have been deposited in the NCBI Gene Expression Omnibus (GEO) and Sequence Read Archive (SRA) under accession no. GSE201470. Mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD043031.

The following data sets were generated
The following previously published data sets were used

Article and author information

Author details

  1. Heon Shin

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-5480-8492
  2. Amy Leung

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
  3. Kevin R Costello

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
  4. Parijat Senapati

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-7324-1230
  5. Hiroyuki Kato

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
  6. Roger E Moore

    Integrated Mass Spectrometry Shared Resource, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
  7. Michael Lee

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
  8. Dimitri Lin

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
  9. Xiaofang Tang

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
  10. Patrick Pirrotte

    Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, United States
    Competing interests
    The authors declare that no competing interests exist.
  11. Zhen Bouman Chen

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-3291-1090
  12. Dustin E Schones

    Department of Diabetes Complications and Metabolism, City of Hope, Duarte, United States
    For correspondence
    dschones@coh.org
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7692-8583

Funding

National Institutes of Health (R01DK112041)

  • Dustin E Schones

National Institutes of Health (R01CA220693)

  • Dustin E Schones

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

Reviewing Editor

  1. Carlos Isales, Augusta University, United States

Ethics

Animal experimentation: All animal experiments conducted have been approved by the Institutional Animal Care and Use Committees at City of Hope. All of the animals were handled according to approved institutional animal care and use committee (IACUC) protocols (#17010).

Version history

  1. Preprint posted: May 11, 2022 (view preprint)
  2. Received: December 15, 2022
  3. Accepted: July 19, 2023
  4. Accepted Manuscript published: July 20, 2023 (version 1)
  5. Version of Record published: July 31, 2023 (version 2)

Copyright

© 2023, Shin 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

  • 1,280
    views
  • 232
    downloads
  • 4
    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. Heon Shin
  2. Amy Leung
  3. Kevin R Costello
  4. Parijat Senapati
  5. Hiroyuki Kato
  6. Roger E Moore
  7. Michael Lee
  8. Dimitri Lin
  9. Xiaofang Tang
  10. Patrick Pirrotte
  11. Zhen Bouman Chen
  12. Dustin E Schones
(2023)
Inhibition of DNMT1 methyltransferase activity via glucose-regulated O-GlcNAcylation alters the epigenome
eLife 12:e85595.
https://doi.org/10.7554/eLife.85595

Share this article

https://doi.org/10.7554/eLife.85595

Further reading

    1. Chromosomes and Gene Expression
    Rupam Choudhury, Anuroop Venkateswaran Venkatasubramani ... Axel Imhof
    Research Article

    Eukaryotic chromatin is organized into functional domains, that are characterized by distinct proteomic compositions and specific nuclear positions. In contrast to cellular organelles surrounded by lipid membranes, the composition of distinct chromatin domains is rather ill described and highly dynamic. To gain molecular insight into these domains and explore their composition, we developed an antibody-based proximity-biotinylation method targeting the RNA and proteins constituents. The method that we termed Antibody-Mediated-Proximity-Labelling-coupled to Mass Spectrometry (AMPL-MS) does not require the expression of fusion proteins and therefore constitutes a versatile and very sensitive method to characterize the composition of chromatin domains based on specific signature proteins or histone modifications. To demonstrate the utility of our approach we used AMPL-MS to characterize the molecular features of the chromocenter as well as the chromosome territory containing the hyperactive X-chromosome in Drosophila. This analysis identified a number of known RNA binding proteins in proximity of the hyperactive X and the centromere, supporting the accuracy of our method. In addition, it enabled us to characterize the role of RNA in the formation of these nuclear bodies. Furthermore, our method identified a new set of RNA molecules associated with the Drosophila centromere. Characterization of these novel molecules suggested the formation of R-loops in centromeres, which we validated using a novel probe for R-loops in Drosophila. Taken together, AMPL-MS improves the selectivity and specificity of proximity ligation allowing for novel discoveries of weak protein-RNA interactions in biologically diverse domains.

    1. Cancer Biology
    2. Chromosomes and Gene Expression
    Gregory Caleb Howard, Jing Wang ... William P Tansey
    Research Article

    The chromatin-associated protein WD Repeat Domain 5 (WDR5) is a promising target for cancer drug discovery, with most efforts blocking an arginine-binding cavity on the protein called the ‘WIN’ site that tethers WDR5 to chromatin. WIN site inhibitors (WINi) are active against multiple cancer cell types in vitro, the most notable of which are those derived from MLL-rearranged (MLLr) leukemias. Peptidomimetic WINi were originally proposed to inhibit MLLr cells via dysregulation of genes connected to hematopoietic stem cell expansion. Our discovery and interrogation of small-molecule WINi, however, revealed that they act in MLLr cell lines to suppress ribosome protein gene (RPG) transcription, induce nucleolar stress, and activate p53. Because there is no precedent for an anticancer strategy that specifically targets RPG expression, we took an integrated multi-omics approach to further interrogate the mechanism of action of WINi in human MLLr cancer cells. We show that WINi induce depletion of the stock of ribosomes, accompanied by a broad yet modest translational choke and changes in alternative mRNA splicing that inactivate the p53 antagonist MDM4. We also show that WINi are synergistic with agents including venetoclax and BET-bromodomain inhibitors. Together, these studies reinforce the concept that WINi are a novel type of ribosome-directed anticancer therapy and provide a resource to support their clinical implementation in MLLr leukemias and other malignancies.