4.7 Article

Long non-coding RNA Meg3 deficiency impairs glucose homeostasis and insulin signaling by inducing cellular senescence of hepatic endothelium in obesity

Journal

REDOX BIOLOGY
Volume 40, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.redox.2021.101863

Keywords

Long noncoding RNAs; Cellular senescence; Obesity; Hepatic endothelium; Glucose homeostasis

Funding

  1. National Institute of General Medical Sciences of the National Institutes of Health [P30GM103335]
  2. National Institutes of Health [P30GM103335, 1P20GM104320, HL150536, HL115141, HL134849, HL148207, HL148355, HL153356, GM108975, GM131701-01, HSN276201200017C]
  3. American Heart Association SDG [15SDG25400012, 18SFRN33900144]
  4. National Institutes of Health Funded T32 grant [5T32GM10700104]
  5. Boston Nutrition Obesity Research Center - NIH grant [P30DK046200]
  6. National Institutes of Health

Ask authors/readers for more resources

The study found that the lncRNA Meg3 plays a role in obesity-induced insulin resistance by inducing cellular senescence of endothelial cells. Knockdown of Meg3 exacerbates the disease progression by promoting cellular senescence in hepatic endothelium.
Obesity-induced insulin resistance is a risk factor for diabetes and cardiovascular disease. However, the mechanisms underlying endothelial senescence in obesity, and how it impacts obesity-induced insulin resistance remain incompletely understood. In this study, transcriptome analysis revealed that the long non-coding RNA (lncRNA) Maternally expressed gene 3 (Meg3) is one of the top differentially expressed lncRNAs in the vascular endothelium in diet-induced obese mice. Meg3 knockdown induces cellular senescence of endothelial cells characterized by increased senescence-associated beta-galactosidase activity, increased levels of endogenous superoxide, impaired mitochondrial structure and function, and impaired autophagy. Moreover, Meg3 knockdown causes cellular senescence of hepatic endothelium in diet-induced obese mice. Furthermore, Meg3 expression is elevated in human nonalcoholic fatty livers and nonalcoholic steatohepatitis livers, which positively correlates with the expression of CDKN2A encoding p16, an important hallmark of cellular senescence. Meg3 knockdown potentiates obesity-induced insulin resistance and impairs glucose homeostasis. Insulin signaling is reduced by Meg3 knockdown in the liver and, to a lesser extent, in the skeletal muscle, but not in the visceral fat of obese mice. We found that the attenuation of cellular senescence of hepatic endothelium by ablating p53 expression in vascular endothelium can restore impaired glucose homeostasis and insulin signaling in obesity. In conclusion, our data demonstrate that cellular senescence of hepatic endothelium promotes obesity-induced insulin resistance, which is tightly regulated by the expression of Meg3. Our results suggest that manipulation of Meg3 expression may represent a novel approach to managing obesity-associated hepatic endothelial senescence and insulin resistance.

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