N-acetyl cysteine prevents arecoline-inhibited C2C12 myoblast differentiation through ERK1/2 phosphorylation

PLoS One. 2022 Jul 28;17(7):e0272231. doi: 10.1371/journal.pone.0272231. eCollection 2022.

Abstract

Arecoline is known to induce reactive oxygen species (ROS). Our previous studies showed that arecoline inhibited myogenic differentiation and acetylcholine receptor cluster formation of C2C12 myoblasts. N-acetyl-cysteine (NAC) is a known ROS scavenger. We hypothesize that NAC scavenges the excess ROS caused by arecoline. In this article we examined the effect of NAC on the inhibited myoblast differentiation by arecoline and related mechanisms. We found that NAC less than 2 mM is non-cytotoxic to C2C12 by viability analysis. We further demonstrated that NAC attenuated the decreased number of myotubes and nuclei in each myotube compared to arecoline treatment by H & E staining. We also showed that NAC prevented the decreased expression level of the myogenic markers, myogenin and MYH caused by arecoline, using immunocytochemistry and western blotting. Finally, we found that NAC restored the decreased expression level of p-ERK1/2 by arecoline. In conclusion, our results indicate that NAC attenuates the damage of the arecoline-inhibited C2C12 myoblast differentiation by the activation/phosphorylation of ERK. This is the first report to demonstrate that NAC has beneficial effects on skeletal muscle myogenesis through ERK1/2 upon arecoline treatment. Since defects of skeletal muscle associates with several diseases, NAC can be a potent drug candidate in diseases related to defects in skeletal muscle myogenesis.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acetylcysteine / metabolism
  • Acetylcysteine / pharmacology
  • Arecoline* / pharmacology
  • Cell Differentiation
  • MAP Kinase Signaling System*
  • Muscle Development
  • Myoblasts / metabolism
  • Phosphorylation
  • Reactive Oxygen Species / metabolism

Substances

  • Reactive Oxygen Species
  • Arecoline
  • Acetylcysteine

Grants and funding

This work was supported in part by grants from The Ministry of Science and Technology, Taiwan to Y.-F.C. (108-2314-B-037-075) and the Kaohsiung Medical University Research Foundation to Y.-F.C. (KMU-M103001, KMU-M104003, KMU-TP104PR16). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.