4.7 Article

Long noncoding RNA SMUL suppresses SMURF2 production-mediated muscle atrophy via nonsense-mediated mRNA decay

Journal

MOLECULAR THERAPY-NUCLEIC ACIDS
Volume 23, Issue -, Pages 512-526

Publisher

CELL PRESS
DOI: 10.1016/j.omtn.2020.12.003

Keywords

-

Funding

  1. Natural Scientific Foundation of China [U1901206, 31802051]
  2. Local Innovative and Research Teams Project of Guangdong Province [2019BT02N630]
  3. TenThousand Talents Program [W03020593]
  4. Chinese Postdoctoral Science Foundation [2017M622715]
  5. Natural Science Foundation of Guangdong Province [2018A030310209]
  6. China Agricultural Research System [CARS41G03]

Ask authors/readers for more resources

104 myogenic-associated lncRNAs were found to be translated into small peptides, and a novel muscle atrophy-associated lncRNA named SMUL was identified, which promoted myoblast proliferation and suppressed differentiation. SMUL restrained SMURF2 production via NMD, participating in the regulation of the TGF-beta/SMAD pathway.
As the world population grows, muscle atrophy leading to muscle wasting could become a bigger risk. Long noncoding RNAs (lncRNAs) are known to play important roles in muscle growth and muscle atrophy. Meanwhile, it has recently come to light that many putative small open reading frames (sORFs) are hidden in lncRNAs; however, their translational capabilities and functions remain unclear. In this study, we uncovered 104 myogenic-associated lncRNAs translated, in at least a small peptide, by integrated transcriptome and proteomic analyses. Furthermore, an upstream ORF (uORF) regulatory network was constructed, and a novel muscle atrophy-associated lncRNA named SMUL (Smad ubiquitin regulatory factor 2 [SMURF2] upstream lncRNA) was identified. SMUL was highly expressed in skeletal muscle, and its expression level was down regulated during myoblast differentiation. SMUL promoted myoblast proliferation and suppressed differentiation in vitro. In vivo, SMUL induced skeletal muscle atrophy and promoted a switch from slow-twitch to fast-twitch fibers. In the meantime, translation of the SMUL sORF disrupted the stability of SMURF2 mRNA. Mechanistically, SMUL restrained SMURF2 production via nonsense-mediated mRNA decay (NMD), participating in the regulation of the transforming growth factor beta (TGF-beta)/SMAD pathway and further regulating myogenesis and muscle atrophy. Taken together, these results suggest that SMUL could be a novel therapeutic target for muscle atrophy.

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