4.8 Article

OLIG2 regulates lncRNAs and its own expression during oligodendrocyte lineage formation

Journal

BMC BIOLOGY
Volume 19, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s12915-021-01057-6

Keywords

Oligodendrocyte development; OLIG2; LncRNAs; Histone modification; Transcriptional regulation; Regulation after transcription

Categories

Funding

  1. National Institutes of Health [R01 NS088353, R21 NS113068-01, R21 EY028647-01]
  2. The Staman Ogilvie Fund-Memorial Hermann Foundation
  3. Mission Connect, a program of The Institute for Rehabilitation and Research (TIRR) Foundation

Ask authors/readers for more resources

The study integrated the genome-wide OLIG2 binding and epigenetic modification status of coding and non-coding genes during different stages of oligodendrocyte differentiation, revealing potential functions and regulation of lncRNAs by OLIG2. It discovered dynamic regulation of bivalent genes by OLIG2 during oligodendrocyte development and a novel self-regulation mechanism of OLIG2 through its own mRNA 3' UTR. These findings provide insights into the transcriptional and post-transcriptional mechanisms underlying oligodendrocyte development and contribute to understanding cell fate decisions during oligodendrocyte lineage formation.
Background Oligodendrocytes, responsible for axon ensheathment, are critical for central nervous system (CNS) development, function, and diseases. OLIG2 is an important transcription factor (TF) that acts during oligodendrocyte development and performs distinct functions at different stages. Previous studies have shown that lncRNAs (long non-coding RNAs; > 200 bp) have important functions during oligodendrocyte development, but their roles have not been systematically characterized and their regulation is not yet clear. Results We performed an integrated study of genome-wide OLIG2 binding and the epigenetic modification status of both coding and non-coding genes during three stages of oligodendrocyte differentiation in vivo: neural stem cells (NSCs), oligodendrocyte progenitor cells (OPCs), and newly formed oligodendrocytes (NFOs). We found that 613 lncRNAs have OLIG2 binding sites and are expressed in at least one cell type, which can potentially be activated or repressed by OLIG2. Forty-eight of them have increased expression in oligodendrocyte lineage cells. Predicting lncRNA functions by using a guilt-by-association approach revealed that the functions of these 48 lncRNAs were enriched in oligodendrocyte development and differentiation. Additionally, bivalent genes are known to play essential roles during embryonic stem cell differentiation. We identified bivalent genes in NSCs, OPCs, and NFOs and found that some bivalent genes bound by OLIG2 are dynamically regulated during oligodendrocyte development. Importantly, we unveiled a previously unknown mechanism that, in addition to transcriptional regulation via DNA binding, OLIG2 could self-regulate through the 3 ' UTR of its own mRNA. Conclusions Our studies have revealed the missing links in the mechanisms regulating oligodendrocyte development at the transcriptional level and after transcription. The results of our research have improved the understanding of fundamental cell fate decisions during oligodendrocyte lineage formation, which can enable insights into demyelination diseases and regenerative medicine.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available