DOI QR코드

DOI QR Code

Effects of alpha-linolenic acid and essential amino acids on the proliferation and differentiation of C2C12 myoblasts

  • Zhou, Dongjie (Department of Animal Science, Chungbuk National University) ;
  • Li, Xiao-Han (Department of Animal Science, Chungbuk National University) ;
  • Lee, Song‑Hee (Department of Animal Science, Chungbuk National University) ;
  • Heo, Geun (Department of Animal Science, Chungbuk National University) ;
  • Cui, Xiang-Shun (Department of Animal Science, Chungbuk National University)
  • Received : 2022.03.07
  • Accepted : 2022.03.17
  • Published : 2022.03.31

Abstract

Alpha-linolenic acid is an important polyunsaturated fatty acid that exhibits anticancer, anti-inflammatory, and antioxidative effects. In this study, we investigated the protective effects of alpha-linolenic acid on the cell proliferation and differentiation of C2C12 cells under essential amino acid-deficient conditions. Different concentrations of alpha-linolenic acid and essential amino acids were added to the growth and differentiation media. The concentrations of 10 µM of alpha-linolenic acid and 2% essential amino acid were chosen for subsequent experiments. Supplementation with alpha-linolenic acid and essential amino acids improved the proliferation and differentiation of C2C12 cells and significantly increased the mRNA levels of catalase, superoxide dismutase, B-cell lymphoma-2, and beclin-1 as well as the protein levels of PPARγ coactivator-1α compared to those in the controls. Moreover, supplementation with alpha-linolenic acid and essential amino acids reduced the levels of phosphorylated H2A.X variant histone, Bcl-2-associated X, p53, and light chain 3 during C2C12 cell proliferation, and increased the expression levels of myogenic factors 4 (myogenin) and 5 during C2C12 cell differentiation. Overall, we determined that alpha-linolenic acid and essential amino acids maintained the cell proliferation and differentiation of C2C12 cells via their anti-oxidative, anti-apoptotic, and anti-autophagic effects.

Keywords

Acknowledgement

This result was supported by "Regional Innovation Strategy (RIS)" through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (MOE) (2021RIS-001).

References

  1. Cho HJ and Kim TM. 2021. CDDO-Me alleviates oxidative stress in human mesenchymal stem cells. J. Anim. Reprod. Biotechnol. 36:285-291. https://doi.org/10.12750/JARB.36.4.285
  2. Daval JL, Pourie G, Grojean S, Lievre V, Strazielle C, Blaise S, Vert P. 2004. Neonatal hypoxia triggers transient apoptosis followed by neurogenesis in the rat CA1 hippocampus. Pediatr. Res. 55:561-567. https://doi.org/10.1203/01.PDR.0000113771.51317.37
  3. Dickinson JM, Gundermann DM, Walker DK, Reidy PT, Borack MS, Drummond MJ, Arora M, Volpi E, Rasmussen BB. 2014. Leucine-enriched amino acid ingestion after resistance exercise prolongs myofibrillar protein synthesis and amino acid transporter expression in older men. J. Nutr. 144:1694-1702. https://doi.org/10.3945/jn.114.198671
  4. Dickinson JM, Volpi E, Rasmussen BB. 2013. Exercise and nutrition to target protein synthesis impairments in aging skeletal muscle. Exerc. Sport Sci. Rev. 41:216-223. https://doi.org/10.1097/JES.0b013e3182a4e699
  5. Fry CS, Drummond MJ, Glynn EL, Dickinson JM, Gundermann DM, Timmerman KL, Walker DK, Dhanani S, Volpi E, Rasmussen BB. 2011. Aging impairs contraction-induced human skeletal muscle mTORC1 signaling and protein synthesis. Skelet. Muscle 1:11. https://doi.org/10.1186/2044-5040-1-11
  6. Guo K and Walsh K. 1997. Inhibition of myogenesis by multiple cyclin-Cdk complexes. Coordinate regulation of myogenesis and cell cycle activity at the level of E2F. J. Biol. Chem. 272:791-797. https://doi.org/10.1074/jbc.272.2.791
  7. Hussain G, Schmitt F, Loeffler JP, Gonzalez de Aguilar JL. 2013. Fatting the brain: a brief of recent research. Front. Cell. Neurosci. 7:144. https://doi.org/10.3389/fncel.2013.00144
  8. Lee HJ, Jeon RH, Park BJ, Jang SJ, Lee SL, Rho GJ, Kim SJ, Lee WJ. 2019. Differentiation inductions altered telomere length and telomerase activity in human dental pulpderived mesenchymal stem cell. J. Anim. Reprod. Biotechnol. 34:93-99. https://doi.org/10.12750/JARB.34.2.93
  9. Leick L, Lyngby SS, Wojtaszewski JF, Pilegaard H. 2010. PGC1alpha is required for training-induced prevention of age-associated decline in mitochondrial enzymes in mouse skeletal muscle. Exp. Gerontol. 45:336-342. (Correcction published 2010, Exp. Gerontol., 45, p. 988) https://doi.org/10.1016/j.exger.2010.01.011
  10. McKinsey TA, Zhang CL, Olson EN. 2001. Control of muscle development by dueling HATs and HDACs. Curr. Opin. Genet. Dev. 11:497-504. https://doi.org/10.1016/S0959-437X(00)00224-0
  11. Park MH, Park JE, Kim MS, Lee KY, Yun JI, Choi JH, Lee E, Lee ST. 2014. Effects of suspension culture on proliferation and undifferentiation of spermatogonial stem cells derived from porcine neonatal testis. Reprod. Dev. Biol. 38:85-91. https://doi.org/10.12749/RDB.2014.38.2.85
  12. Porrello A, Cerone MA, Coen S, Gurtner A, Fontemaggi G, Cimino L, Piaggio G, Sacchi A, Soddu S. 2000. p53 regulates myogenesis by triggering the differentiation activity of pRb. J. Cell Biol. 151:1295-1304. https://doi.org/10.1083/jcb.151.6.1295
  13. Qu A, Jiang C, Xu M, Zhang Y, Zhu Y, Xu Q, Zhang C, Wang X. 2009. PGC-1alpha attenuates neointimal formation via inhibition of vascular smooth muscle cell migration in the injured rat carotid artery. Am. J. Physiol. Cell Physiol. 297:C645-C653. https://doi.org/10.1152/ajpcell.00469.2008
  14. Rescan PY. 2001. Regulation and functions of myogenic regulatory factors in lower vertebrates. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 130:1-12. https://doi.org/10.1016/S1096-4959(01)00412-2
  15. Roqueta-Rivera M, Abbott TL, Sivaguru M, Hess RA, Nakamura MT. 2011. Deficiency in the omega-3 fatty acid pathway results in failure of acrosome biogenesis in mice. Biol. Reprod. 85:721-732. https://doi.org/10.1095/biolreprod.110.089524
  16. Roy S, Rawat AK, Sammi SR, Devi U, Singh M, Gautam S, Yadav RK, Rawat JK, Singh L, Ansari MN, Saeedan AS, Pandey R, Kumar D, Kaithwas G. 2017. Alpha-linolenic acid stabilizes HIF-1 α and downregulates FASN to promote mitochondrial apoptosis for mammary gland chemoprevention. Oncotarget 8:70049-70071. https://doi.org/10.18632/oncotarget.19551
  17. Shen L, Sun B, Sheng J, Yu S, Li Y, Xu H, Su J, Sun L. 2018. PGC1α promotes cisplatin resistance in human ovarian carcinoma cells through upregulation of mitochondrial biogenesis. Int. J. Oncol. 53:404-416.
  18. Skapek SX, Rhee J, Kim PS, Novitch BG, Lassar AB. 1996. Cyclin-mediated inhibition of muscle gene expression via a mechanism that is independent of pRB hyperphosphorylation. Mol. Cell. Biol. 16:7043-7053. https://doi.org/10.1128/MCB.16.12.7043
  19. Valente LJ, Gray DH, Michalak EM, Pinon-Hofbauer J, Egle A, Scott CL, Janic A, Strasser A. 2013. p53 efficiently suppresses tumor development in the complete absence of its cell-cycle inhibitory and proapoptotic effectors p21, Puma, and Noxa. Cell Rep. 3:1339-1345. https://doi.org/10.1016/j.celrep.2013.04.012
  20. Walker DK, Dickinson JM, Timmerman KL, Drummond MJ, Reidy PT, Fry CS, Gundermann DM, Rasmussen BB. 2011. Exercise, amino acids, and aging in the control of human muscle protein synthesis. Med. Sci. Sports Exerc. 43:2249-2258. https://doi.org/10.1249/MSS.0b013e318223b037
  21. Wang X, Breeze A, Kulka M. 2015. N-3 polyunsaturated fatty acids inhibit IFN-γ-induced IL-18 binding protein production by prostate cancer cells. Cancer Immunol. Immunother. 64:249-258. https://doi.org/10.1007/s00262-014-1630-z
  22. Wathes DC, Abayasekara DR, Aitken RJ. 2007. Polyunsaturated fatty acids in male and female reproduction. Biol. Reprod. 77:190-201. https://doi.org/10.1095/biolreprod.107.060558
  23. Xia Z, Dickens M, Raingeaud J, Davis RJ, Greenberg ME. 1995. Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science 270:1326-1331. https://doi.org/10.1126/science.270.5240.1326