Skip to main content
Log in

Magnolol inhibits myotube atrophy induced by cancer cachexia through myostatin signaling pathway in vitro

  • Original Paper
  • Published:
Journal of Natural Medicines Aims and scope Submit manuscript

Abstract

Cancer cachexia is a complex and multifactorial syndrome that influences about 50–80% of cancer patients and may lead to 20% of cancer deaths and muscle atrophy is the key characteristic of the syndrome. Recent researches have shown that myostatin is a negative regulator in the growth and differentiation of skeletal muscle. Herein, C2C12 cancer cachexia model was established with C26 conditioned culture medium (CCM), then treated with magnolol to evaluate the pharmacological activity of magnolol in myotube atrophy. Our results demonstrated that magnolol inhibited the activity of myostatin promotor and the myostatin signaling pathway. In C2C12 cancer cachexia model, magnolol decreased myostatin expression, inhibited the phosphorylation of SMAD2/3 activated by C26 conditioned culture medium (CCM), and elevated the phosphorylation of FOXO3a lowered by CCM. Myosin heavy chain (MyHC), myogenin (MyoG), and myogenic differentiation (MyoD), as three common myotube markers in C2C12 myotube, were decreased by CCM, which could be effectively reversed by magnolol via activation of AKT/mTOR-regulated protein synthesis and inhibition of ubiquitin-mediated proteolysis. This study reveals that magnolol inhibits myotube atrophy induced by CCM by increasing protein synthesis and decreasing ubiquitin-mediated proteolysis, so that magnolol is a promising leading compound in treating muscle atrophy induced by cancer cachexia.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, Jatoi A, Loprinzi C, MacDonald N, Mantovani G, Davis M, Muscaritoli M, Ottery F, Radbruch L, Ravasco P, Walsh D, Wilcock A, Kaasa S, Baracos VE (2011) Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 12:489–495

    Article  Google Scholar 

  2. Kazemi-Bajestani SM, Mazurak VC, Baracos V (2016) Computed tomography-defined muscle and fat wasting are associated with cancer clinical outcomes. Semin Cell Dev Biol 54:2–10

    Article  Google Scholar 

  3. Fearon K, Arends J, Baracos V (2013) Understanding the mechanisms and treatment options in cancer cachexia. Nature reviews Nat Rev Clin Oncol 10:90–99

    Article  CAS  Google Scholar 

  4. Argilés JM, Busquets S, Stemmler B, López-Soriano FJ (2014) Cancer cachexia: understanding the molecular basis. Nat Rev Cancer 14:754–762

    Article  Google Scholar 

  5. Argilés JM, López-Soriano FJ (1996) The ubiquitin-dependent proteolytic pathway in skeletal muscle: its role in pathological states. Trends Pharmacol Sci 17:223–226

    Article  Google Scholar 

  6. Gomes MD, Lecker SH, Jagoe RT, Navon A, Goldberg AL (2001) Atrogin-1, A Muscle-Specific F-Box protein highly expressed during muscle atrophy. Proc Natl Acad Sci USA 98:14440–14445

    Article  CAS  Google Scholar 

  7. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA, Poueymirou WT, Panaro FJ, Na E, Dharmarajan K, Pan ZQ, Valenzuela DM, DeChiara TM, Stitt TN, Yancopoulos GD, Glass DJ (2001) Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 294:1704–1708

    Article  CAS  Google Scholar 

  8. Rodriguez J, Vernus B, Chelh I (2014) Myostatin and the skeletal muscle atrophy and hypertrophy signaling pathways. Cell Mol Life Sci 71:4361–4371

    Article  CAS  Google Scholar 

  9. Cohen S, Nathan JA, Goldberg AL (2015) Muscle wasting in disease: molecular mechanisms and promising therapies. Nat Rev Drug Discov 14:58–74

    Article  CAS  Google Scholar 

  10. Han HQ, Zhou X, Mitch WE, Goldberg AL (2013) Myostatin/activin pathway antagonism: Molecular basis and therapeutic potential. Int J Biochem Cell Biol 45:2333–2347

    Article  CAS  Google Scholar 

  11. Murphy KT, Chee A, Gleeson BG, Naim T, Swiderski K, Koopman R, Lynch GS (2011) Antibody-directed myostatin inhibition enhances muscle mass and function in tumor-bearing mice. Am J Physiol Regul Integr Comp Physiol 301:R716–R726

    Article  CAS  Google Scholar 

  12. Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, Rosenfeld R, Chen Q, Boone T, Simonet WS, Lacey DL, Goldberg AL, Han HQ (2010) Reversal of cancer cachexia and muscle wasting by actriib antagonism leads to prolonged survival. Cell 142:531–543

    Article  CAS  Google Scholar 

  13. Hoogaars WMH, Jaspers RT (2018) Past, present, and future perspective of targeting myostatin and related signaling pathways to counteract muscle atrophy. Adv Exp Med Biol 1088:153–206

    Article  CAS  Google Scholar 

  14. Lee DH, Szczepanski MJ, Lee YJ (2009) Magnolol induces apoptosis via inhibiting the EGFR/PI3K/Akt signaling pathway in human prostate cancer cells. J Cell Biochem 106:1113–1122

    Article  CAS  Google Scholar 

  15. Ranaware AM, Banik K, Deshpande V, Padmavathi G, Roy NK, Sethi G, Fan L, Kumar AP, Kunnumakkara AB (2018) Magnolol: a neolignan from the magnolia family for the prevention and treatment of cancer. Int J Mol Sci 19:E2362

    Article  Google Scholar 

  16. Liu D, Qiao X, Ge Z et al (2019) IMB0901 inhibits muscle atrophy induced by cancer cachexia through MSTN signaling pathway. Skelet Muscle 9:8

    Article  Google Scholar 

  17. Thomas M, Langley B, Berry C, Sharma M, Kirk S, Bass J, Kambadur R (2000) Myostatin, a negative regulator of muscle growth, functions by inhibiting myoblast proliferation. J Biol Chem 275:40235–40243

    Article  CAS  Google Scholar 

  18. Cash JN, Angerman EB, Kirby RJ, Merck L, Seibel WL, Wortman MD, Papoian R, Nelson S, Thompson TB (2013) Development of a small-molecule screening method for inhibitors of cellular response to myostatin and activin a. J Biomol Screen 18:837–844

    Article  CAS  Google Scholar 

  19. Liao K, Yong CW, Hua K (2018) SB431542 inhibited cigarette smoke extract induced invasiveness of A549 cells via the TGF-β1/Smad2/MMP3 pathway. Oncol Lett 15:9681–9686

    PubMed  PubMed Central  Google Scholar 

  20. Benny Klimek ME, Aydogdu T, Link MJ, Pons M, Koniaris LG, Zimmers TA (2010) Acute inhibition of myostatin-family proteins preserves skeletal muscle in mouse models of cancer cachexia. Biochem Biophys Res Commun 391:1548–1554

    Article  CAS  Google Scholar 

  21. Gallot YS, Durieux AC, Castells J, Desgeorges MM, Vernus B, Plantureux L, Rémond D, Jahnke VE, Lefai E, Dardevet D, Nemoz G, Schaeffer L, Bonnieu A, Freyssenet DG (2014) Myostatin gene inactivation prevents skeletal muscle wasting in cancer. Can Res 74:7344–7356

    Article  CAS  Google Scholar 

  22. Sartori R, Milan G, Patron M, Mammucari C, Blaauw B, Abraham R, Sandri M (2009) Smad2 and 3 transcription factors control muscle mass in adulthood. Am J Physiol Cell Physiol 296:C1248–C1257

    Article  CAS  Google Scholar 

  23. Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ, Yancopoulos GD (2001) Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol 3:1014–1019

    Article  CAS  Google Scholar 

  24. Song SE, Shin SK, Park SY, Hwang IS, Im SS, Bae JH, Choi MS, Song DK (2018) Epac2a-knockout mice are resistant to dexamethasone-induced skeletal muscle atrophy and short-term cold stress. BMB Rep 51:39–44

    Article  CAS  Google Scholar 

  25. Trendelenburg AU, Meyer A, Rohner D, Boyle J, Hatakeyama S, Glass DJ (2009) Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am J Physiol Cell Physiol 296:C1258–C1270

    Article  CAS  Google Scholar 

  26. Milan G, Romanello V, Pescatore F, Armani A, Paik JH, Frasson L, Seydel A, Zhao J, Abraham R, Goldberg AL, Blaauw B, DePinho RA, Sandri M (2015) Regulation of autophagy and the ubiquitin-proteasome system by the FoxO transcriptional network during muscle atrophy. Nat Commun 6:6670

    Article  CAS  Google Scholar 

  27. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schiaffino S, Lecker SH, Goldberg AL (2004) Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117:399–412

    Article  CAS  Google Scholar 

  28. Lokireddy S, Mouly V, Butler-Browne G, Gluckman PD, Sharma M, Kambadur R, McFarlane C (2011) Myostatin promotes the wasting of human myoblast cultures through promoting ubiquitin-proteasome pathway-mediated loss of sarcomeric proteins. Am J Physiol Cell Physiol 301:1316–1324

    Article  Google Scholar 

  29. Zammit PS (2017) Function of the myogenic regulatory factors Myf5, MyoD, Myogenin and MRF4 in skeletal muscle, satellite cells and regenerative myogenesis. Semin Cell Dev Biol 72:19–32

    Article  CAS  Google Scholar 

  30. McFarlane C, Hui GZ, Amanda WZ, Lau HY, Lokireddy S, Xiaojia G, Mouly V, Butler-Browne G, Gluckman PD, Sharma M, Kambadur R (2011) Human myostatin negatively regulates human myoblast growth and differentiation. Am J Physiol Cell Physiol 301:195–203

    Article  Google Scholar 

  31. Langley B, Thomas M, Bishop A, Sharma M, Gilmour S, Kambadur R (2002) Myostatin inhibits myoblast differentiation by down-regulating myod expression. J Biol Chem 277:49831–49840

    Article  CAS  Google Scholar 

  32. Golan T, Geva R, Richards D, Madhusudan S, Lin BK, Wang HT, Walgren RA, Stemmer SM (2018) LY2495655, an antimyostatin antibody, in pancreatic cancer: a randomized, phase 2 trial. J Cachexia Sarcopenia Muscle 9:871–879

    Article  Google Scholar 

Download references

Acknowledgments

This work was supported by grants from CAMS Innovation Fund for Medical Sciences (CIFMS, 2016-I2M-02-002) and “Significant New Drug Development” Major Science and Technology Development Projects of China (No.2018ZX09711001-007-002).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shu-zhen Chen.

Ethics declarations

Conflicts of interest

The authors have no conflicting interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ge, Z., Liu, D., Shang, Y. et al. Magnolol inhibits myotube atrophy induced by cancer cachexia through myostatin signaling pathway in vitro. J Nat Med 74, 741–749 (2020). https://doi.org/10.1007/s11418-020-01428-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11418-020-01428-3

Keywords

Navigation