4.7 Article

Dynamic Alteration Profile and New Role of RNA m6A Methylation in Replicative and H2O2-Induced Premature Senescence of Human Embryonic Lung Fibroblasts

Journal

Publisher

MDPI
DOI: 10.3390/ijms23169271

Keywords

oxidative stress; premature senescence; m6A; RNA methylation

Funding

  1. Natural Science Foundation of Guangdong Province of China [2021A1515011220]
  2. National Natural Science Foundation of China [81473014]
  3. Outstanding Young Talent of Double Hundred Talents Plan in Jinan University
  4. Open Fund of State Key Laboratory of Respiratory Diseases of China [SKLRD-OP-201810]

Ask authors/readers for more resources

N6-methyladenosine (m6A) methylation plays an important role in cellular senescence. ROS level differs between replicative and premature senescence, leading to changes in the expression profiles of m6A-related enzymes and binding proteins. Total m6A content and RNA methylation enzyme activity decrease in senescent cells. Specific m6A methylation levels regulate the expression of certain genes in replicative and premature senescence.
N6-methyladenosine (m6A) methylation is one of the most common RNA modifications, regulating RNA fate at the posttranscriptional level, and is closely related to cellular senescence. Both models of replicative and premature senescence induced by hydrogen peroxide (H2O2) were used to detect m6A regulation during the senescence of human embryonic lung fibroblasts (HEFs). The ROS level accumulated gradually with senescence, leading to normal replicative senescence. H2O2-treated cells had dramatically increased ROS level, inducing the onset of acute premature senescence. Compared with replicative senescence, ROS changed the expression profiles for m6A-related enzymes and binding proteins, including higher levels of METTL3, METTL14, WTAP, KIAA1429, and FTO, and lower levels of METTL16, ALKBH5, YTHDC1, and YTHDF1/2/3 in the premature senescence persistence group, respectively. Meanwhile, senescent cells decreased total m6A content and RNA methylation enzymes activity, regardless of replicative or premature senescence. Moreover, specific m6A methylation levels regulated the expression of SIRT3, IRS2, and E2F3 between replicative and premature senescence separately. Taken together, differential m6A epitranscription microenvironment and the targeted genes can be used as epigenetic biomarkers to cell senescence and the related diseases, offering new clues for the prevention and intervention of cellular senescence.

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