4.7 Article

Whole-genome profiling of DNA methylation and hydroxymethylation identifies distinct regulatory programs among innate lymphocytes

Journal

NATURE IMMUNOLOGY
Volume 23, Issue 4, Pages 619-+

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s41590-022-01164-8

Keywords

-

Categories

Funding

  1. NIH [F30 DK127540-01, T32 DK 77653-28, R00 DK118110, R21 AI156411, R01HG007175, U24ES026699, U01HG009391, U24HG012070, R01 AI134035, R01 DE025884, 1R01 AI134236-01, R01 DK124699]

Ask authors/readers for more resources

This study characterizes the whole-genome distribution of DNA methylation and hydroxymethylation in innate lymphocytes. It identifies differentially methylated and hydroxymethylated regions between NK cells, ILC2s, and ILC3s and correlates them with transcriptional signatures. The study also reveals unique patterns of DNA methylation/hydroxymethylation in relation to open chromatin regions, histone modifications, and TF-binding sites.
Innate lymphocytes encompass a diverse array of phenotypic identities with specialized functions. DNA methylation and hydroxymethylation are essential for epigenetic fidelity and fate commitment. The landscapes of these modifications are unknown in innate lymphocytes. Here, we characterized the whole-genome distribution of methyl-CpG and 5-hydroxymethylcytosine (5hmC) in mouse innate lymphoid cell 3 (ILC3), ILC2 and natural killer (NK) cells. We identified differentially methylated regions (DMRs) and differentially hydroxymethylated regions (DHMRs) between ILC and NK cell subsets and correlated them with transcriptional signatures. We associated lineage-determining transcription factors (LDTFs) with demethylation and demonstrated unique patterns of DNA methylation/hydroxymethylation in relationship to open chromatin regions (OCRs), histone modifications and TF-binding sites. We further identified an association between hydroxymethylation and NK cell superenhancers (SEs). Using mice lacking the DNA hydroxymethylase TET2, we showed the requirement for TET2 in optimal production of hallmark cytokines by ILC3s and interleukin-17A (IL-17A) by inflammatory ILC2s. These findings provide a powerful resource for studying innate lymphocyte epigenetic regulation and decode the regulatory logic governing their identity. Colonna and colleagues present a genome-wide characterization of DNA methylation and hydroxymethylation in innate lymphocytes and identify differentially methylated and hydroxymethylated regions between NK cells, ILC2s and ILC3s.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available