DOI QR코드

DOI QR Code

Fatty acid oxidation regulates cellular senescence by modulating the autophagy-SIRT1 axis

  • Seungyeon Yang (Department of Biochemistry, College of Medicine, The Catholic University of Korea) ;
  • Subin Moon (Department of Biochemistry, College of Medicine, The Catholic University of Korea) ;
  • Soojung Claire Hur (Department of Mechanical Engineering, Johns Hopkins University) ;
  • Seung Min Jeong (Department of Biochemistry, College of Medicine, The Catholic University of Korea)
  • Received : 2023.05.13
  • Accepted : 2023.09.07
  • Published : 2023.12.31

Abstract

Senescence, a cellular process through which damaged or dysfunctional cells suppress the cell cycle, contributes to aging or age-related functional decline. Cell metabolism has been closely correlated with aging processes, and it has been widely recognized that metabolic changes underlie the cellular alterations that occur with aging. Here, we report that fatty acid oxidation (FAO) serves as a critical regulator of cellular senescence and uncover the underlying mechanism by which FAO inhibition induces senescence. Pharmacological or genetic ablation of FAO results in a p53-dependent induction of cellular senescence in human fibroblasts, whereas enhancing FAO suppresses replicative senescence. We found that FAO inhibition promotes cellular senescence through acetyl-CoA, independent of energy depletion. Mechanistically, increased formation of autophagosomes following FAO inhibition leads to a reduction in SIRT1 protein levels, thereby contributing to senescence induction. Finally, we found that inhibition of autophagy or enforced expression of SIRT1 can rescue the induction of senescence as a result of FAO inhibition. Collectively, our study reveals a distinctive role for the FAO-autophagy-SIRT1 axis in the regulation of cellular senescence.

Keywords

Acknowledgement

This research was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (2019R1A2C1089937 and 2022R1F1A1066394).

References

  1. Guo J, Huang X, Dou L et al (2022) Aging and agingrelated diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther 7, 391
  2. Campisi J and d'Adda di Fagagna F (2007) Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol 8, 729-740 https://doi.org/10.1038/nrm2233
  3. Collado M and Serrano M (2010) Senescence in tumours: evidence from mice and humans. Nat Rev Cancer 10, 51-57 https://doi.org/10.1038/nrc2772
  4. Majumder PK, Grisanzio C, O'Connell F et al (2008) A prostatic intraepithelial neoplasia-dependent p27 Kip1 checkpoint induces senescence and inhibits cell proliferation and cancer progression. Cancer Cell 14, 146-155 https://doi.org/10.1016/j.ccr.2008.06.002
  5. Schosserer M (2022) The role and biology of senescent cells in ageing-related tissue damage and repair. Mech Ageing Dev 202, 111629
  6. van Deursen JM (2014) The role of senescent cells in ageing. Nature 509, 439-446 https://doi.org/10.1038/nature13193
  7. von Kobbe C (2019) Targeting senescent cells: approaches, opportunities, challenges. Aging (Albany NY) 11, 12844-12861 https://doi.org/10.18632/aging.102557
  8. Wiley CD and Campisi J (2021) The metabolic roots of senescence: mechanisms and opportunities for intervention. Nat Metab 3, 1290-1301 https://doi.org/10.1038/s42255-021-00483-8
  9. Catic A (2018) Cellular Metabolism and Aging. Prog Mol Biol Transl Sci 155, 85-107 https://doi.org/10.1016/bs.pmbts.2017.12.003
  10. Salminen A and Kaarniranta K (2012) AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res Rev 11, 230-241 https://doi.org/10.1016/j.arr.2011.12.005
  11. Weichhart T (2018) mTOR as regulator of lifespan, aging, and cellular senescence: a mini-review. Gerontology 64, 127-134 https://doi.org/10.1159/000484629
  12. Toth MJ and Tchernof A (2000) Lipid metabolism in the elderly. Eur J Clin Nutr 54 Suppl 3, S121-125 https://doi.org/10.1038/sj.ejcn.1601033
  13. Chung KW (2021) Advances in understanding of the role of lipid metabolism in aging. Cells 10, 880
  14. Lee G, Kim YY, Jang H et al (2022) SREBP1c-PARP1 axis tunes anti-senescence activity of adipocytes and ameliorates metabolic imbalance in obesity. Cell Metab 34, 702-718 e705
  15. Houten SM, Violante S, Ventura FV and Wanders RJ (2016) The biochemistry and physiology of mitochondrial fatty acid beta-oxidation and its genetic disorders. Annu Rev Physiol 78, 23-44 https://doi.org/10.1146/annurev-physiol-021115-105045
  16. Ma Y, Temkin SM, Hawkridge AM et al (2018) Fatty acid oxidation: an emerging facet of metabolic transformation in cancer. Cancer Lett 435, 92-100 https://doi.org/10.1016/j.canlet.2018.08.006
  17. Carracedo A, Cantley LC and Pandolfi PP (2013) Cancer metabolism: fatty acid oxidation in the limelight. Nat Rev Cancer 13, 227-232 https://doi.org/10.1038/nrc3483
  18. Pougovkina O, te Brinke H, Ofman R et al (2014) Mitochondrial protein acetylation is driven by acetyl-CoA from fatty acid oxidation. Hum Mol Genet 23, 3513-3522 https://doi.org/10.1093/hmg/ddu059
  19. McDonnell E, Crown SB, Fox DB et al (2016) Lipids reprogram metabolism to become a major carbon source for histone acetylation. Cell Rep 17, 1463-1472 https://doi.org/10.1016/j.celrep.2016.10.012
  20. Kumari R and Jat P (2021) Mechanisms of cellular senescence: cell cycle arrest and senescence associated secretory phenotype. Front Cell Dev Biol 9, 645593
  21. Houten SM and Wanders RJ (2010) A general introduction to the biochemistry of mitochondrial fatty acid beta-oxidation. J Inherit Metab Dis 33, 469-477 https://doi.org/10.1007/s10545-010-9061-2
  22. Kim B, Gwak J, Kim M et al (2023) Suppression of fatty acid oxidation supports pancreatic cancer growth and survival under hypoxic conditions through autophagy induction. Cancer Gene Ther 30, 878-889 https://doi.org/10.1038/s41417-023-00598-y
  23. Xu C, Wang L, Fozouni P et al (2020) SIRT1 is downregulated by autophagy in senescence and ageing. Nat Cell Biol 22, 1170-1179 https://doi.org/10.1038/s41556-020-00579-5
  24. Pouikli A, Parekh S, Maleszewska M et al (2021) Chromatin remodeling due to degradation of citrate carrier impairs osteogenesis of aged mesenchymal stem cells. Nature Aging 1, 810-825 https://doi.org/10.1038/s43587-021-00105-8
  25. Bruss MD, Khambatta CF, Ruby MA, Aggarwal I and Hellerstein MK (2010) Calorie restriction increases fatty acid synthesis and whole body fat oxidation rates. Am J Physiol Endocrinol Metab 298, E108-116 https://doi.org/10.1152/ajpendo.00524.2009
  26. Purdom T, Kravitz L, Dokladny K and Mermier C (2018) Understanding the factors that effect maximal fat oxidation. J Int Soc Sports Nutr 15, 3
  27. Serra D, Mera P, Malandrino MI, Mir JF and Herrero L (2013) Mitochondrial fatty acid oxidation in obesity. Antioxid Redox Signal 19, 269-284 https://doi.org/10.1089/ars.2012.4875
  28. Tchkonia T, Morbeck DE, Von Zglinicki T et al (2010) Fat tissue, aging, and cellular senescence. Aging Cell 9, 667-684 https://doi.org/10.1111/j.1474-9726.2010.00608.x
  29. Yang S, Hwang S, Kim M, Seo SB, Lee JH and Jeong SM (2018) Mitochondrial glutamine metabolism via GOT2 supports pancreatic cancer growth through senescence inhibition. Cell Death Dis 9, 55