DOI QR코드

DOI QR Code

Superoxide dismutase 3 protects mesenchymal stem cells through enhanced autophagy and regulation of FoxO3a trafficking

  • Agrahari, Gaurav (Laboratory of Dermato-Immunology, College of Medicine, The Catholic University of Korea) ;
  • Sah, Shyam Kishor (Laboratory of Dermato-Immunology, College of Medicine, The Catholic University of Korea) ;
  • Kim, Tae-Yoon (Laboratory of Dermato-Immunology, College of Medicine, The Catholic University of Korea)
  • Received : 2018.04.11
  • Accepted : 2018.05.24
  • Published : 2018.07.31

Abstract

Therapeutic applications of mesenchymal stem cells (MSCs) are limited due to their early death within the first few days of transplantation. Therefore, to improve the efficacy of cell-based therapies, it is necessary to manipulate MSCs so that they can resist various stresses imposed by the microenvironment. Moreover, the role of superoxide dismutase 3 (SOD3) in regulating such survival under different stress conditions remain elusive. In this study, we overexpressed SOD3 in MSCs (SOD3-MSCs) and evaluated its effect under serum starvation conditions. Nutritional limitation can decrease the survival rate of transplanted MSCs and thus can reduce their efficacy during therapy. Interestingly, we found that SOD3-MSCs exhibited reduced reactive oxygen species levels and greater survival rates than normal MSCs under serum-deprived conditions. In addition, overexpression of SOD3 attenuated starvation-induced apoptosis with increased autophagy in MSCs. Moreover, we have demonstrated that SOD3 protects MSCs against the negative effects of serum deprivation via modulation of AMP-activated protein kinase/sirtulin 1, extracellular signal-regulated kinase activation, and promoted Forkhead box O3a trafficking to the nucleus. Taken together, these results demonstrate that SOD3 promotes MSCs survival and add further evidence to the concept that SOD3-MSCs may be a potential therapeutic agent with better outcomes than normal MSCs for various diseases involving oxidative stress and compromised MSCs survival during therapy.

Keywords

References

  1. Biffi A, Montini E, Lorioli L et al (2013) Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science 341, 1233158 https://doi.org/10.1126/science.1233158
  2. Garbern JC and Lee RT (2013) Cardiac stem cell therapy and the promise of heart regeneration cell. Stem Cell 12, 689-698
  3. Greco SJ and Rameshwar P (2012) Mesenchymal stem cells in drug/gene delivery: implications for cell therapy. TherDeliv 3, 997-1004
  4. Law S and Chaudhuri S (2013) Mesenchymal stem cell and regenerative medicine: regeneration versus immunomodulatory challenges. Am J Stem Cells 2, 22-38
  5. Murphy MB, Moncivais K and Caplan AI (2013) Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med 45, 54 https://doi.org/10.1038/emm.2013.94
  6. Przybyla LM, Theunissen TW, Jaenisch R and Voldman J (2013) Matrix remodeling maintains embryonic stem cell self-renewal by activating stat3. Stem Cells 31, 1097-1106 https://doi.org/10.1002/stem.1360
  7. Saunders A, Faiola F and Wang J (2013) Concise review: pursuing self-renewal and pluripotency with the stem cell factor Nanog. Stem Cells 31, 1227-1236 https://doi.org/10.1002/stem.1384
  8. Haider H and Ashraf M (2010) Preconditioning and stem cell survival. J Cardiovasc Transl Res 3, 89-102 https://doi.org/10.1007/s12265-009-9161-2
  9. Zhou H, Yang J, Xin T et al (2014) Exendin-4 protects adipose-derived mesenchymal stem cells from apoptosis induced by hydrogen peroxide through the PI3K/Akt-Sfrp2 pathways. Free Radic Biol Med 77, 363-375 https://doi.org/10.1016/j.freeradbiomed.2014.09.033
  10. Li CJ, Sun LY and Pang CY (2015) Synergistic protection of N-acetylcysteine and ascorbic acid 2-phosphate on human mesenchymal stem cells against mitoptosis, necroptosis and apoptosis. Sci Rep 5, 9819 https://doi.org/10.1038/srep09819
  11. Zhang Q, Liu S, Li T et al (2016) Preconditioning of bone marrow mesenchymal stem cells with hydrogen sulfide improves their therapeutic potential. Oncotarget 7, 58089-58104
  12. Zhu W, Chen J, Cong X, Hu S and Chen X (2006) Hypoxia and serum deprivation-induced apoptosis in mesenchymal stem cells. Stem Cells 24, 416-425 https://doi.org/10.1634/stemcells.2005-0121
  13. Potier E, Ferreira E, Meunier A, Sedel L, Logeart-Avramoglou D and Petite H (2007) Prolonged hypoxia concomitant with serum deprivation induces massive human mesenchymal stem cell death. Tissue Eng 13, 1325-1331 https://doi.org/10.1089/ten.2006.0325
  14. Mizushima N and Klionsky DJ (2007) Protein turnover via autophagy: implications for metabolism. Annu Rev Nutr 27, 19-40 https://doi.org/10.1146/annurev.nutr.27.061406.093749
  15. Stolz A, Ernst A and Dikic I (2014) Cargo recognition and trafficking in selective autophagy. Nat Cell Biol 16, 495-501 https://doi.org/10.1038/ncb2979
  16. Sbrana FV, Cortini M, Avnet S, Perut F, Columbaro M, De Milito A and Baldini N (2016) The Role of Autophagy in the Maintenance of Stemness and Differentiation of Mesenchymal Stem Cells. Stem Cell Rev 12, 621-633 https://doi.org/10.1007/s12015-016-9690-4
  17. An Y, Liu WJ, Xue P et al (2018) Autophagy promotes MSC-mediated vascularization in cutaneous wound healing via regulation of VEGF secretion. Cell Death Dis 9, 58 https://doi.org/10.1038/s41419-017-0082-8
  18. Gao L, Cen S, Wang P et al (2018) Autophagy Improves the Immunosuppression of $CD4^{+}$ T Cells by Mesenchymal Stem Cells Through Transforming Growth Factor-${\beta}1$. Stem Cells Transl Med 5, 1496-1505
  19. Rubinsztein DC, Bento CF and Deretic V (2015) Therapeutic targeting of autophagy in neurodegenerative and infectious diseases. J Exp Med 212, 979-990 https://doi.org/10.1084/jem.20150956
  20. Zeng W, Xiao J, Zheng G et al (2015) Antioxidant treatment enhances human mesenchymal stem cell anti-stress ability and therapeutic efficacy in an acute liver failure model. Sci Rep 5, 11100 https://doi.org/10.1038/srep11100
  21. Kim PH, Na SS, Lee B, Kim JH and Cho JY (2015) Stanninocalcin 2 enhances mesenchymal stem cell survival by suppressing oxidaative stress. BMB Rep 48, 702-707 https://doi.org/10.5483/BMBRep.2015.48.12.158
  22. Suh N and Lee Eb (2017) Antioxidant effects of selenocysteine on replicative senescence in human adipose-derived mesenchymal stem cells. BMB Rep 50, 572-577 https://doi.org/10.5483/BMBRep.2017.50.11.174
  23. Kemp K, Hares K, Mallam E, Heesom KJ, Scolding N and Wilkins A (2010) Mesenchymal stem cell-secreted superoxide dismutase promotes cerebellar neuronal survival. J Neurochem 114, 1569-1580 https://doi.org/10.1111/j.1471-4159.2009.06553.x
  24. Egan DF, Shackelford DB, Mihaylova MM et al (2011) Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331, 456-461 https://doi.org/10.1126/science.1196371
  25. Kim J, Kundu M, Viollet B and Guan KL (2011) AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 13, 132-141 https://doi.org/10.1038/ncb2152
  26. Laplante M and Sabatini DM (2012) mTOR signaling in growth control and disease. Cell 149, 274-293 https://doi.org/10.1016/j.cell.2012.03.017
  27. Salih DA and Brunet A (2008) FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol 20, 126-136 https://doi.org/10.1016/j.ceb.2008.02.005
  28. Wang j, Whiteman MW, Lian H et al (2009) A Non-canonical MEK/ERK Signaling Pathway Regulates Autophagy via Regulating Beclin 1. J Biol Chem 284, 21412-21424 https://doi.org/10.1074/jbc.M109.026013
  29. Lee SB, Kim JJ, Kim TW, Kim BS, Lee MS and Yoo YD (2010) Serum deprivation induced reactive oxygen species production is mediated by Romo1. Apoptosis 15, 204-218 https://doi.org/10.1007/s10495-009-0411-1
  30. Richard CW, Yongjie W, Zhenyi A et al (2012) Akt-Mediated Regulation of Autophagy and Tumorigenesis Through Beclin 1 Phosphorylation. Science 338, 956-959 https://doi.org/10.1126/science.1225967
  31. Mammucari C, Milan G, Romanello V et al (2007) FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6, 458-471 https://doi.org/10.1016/j.cmet.2007.11.001
  32. Mammucari C, Schiaffino S and Sandri M (2008) Downstream of Akt: FoxO3 and mTOR in the regulation of autophagy in skeletal muscle. Autophagy 4, 524-526 https://doi.org/10.4161/auto.5905
  33. Van der Vos KE, Eliasson P, Proikas-Cezanne T et al (2012) Modulation of glutamine metabolism by the PI(3)K-PKB-FOXO network regulates autophagy. Nat Cell Biol 14, 829-837 https://doi.org/10.1038/ncb2536
  34. Xiong X, Tao R, Depinho R and Charlie Dong X (2012) The autophagy-related gene 14 (Atg14) is regulated by Forkhead box O transcription factors and circadian rhythms and plays a critical role in hepatic autophagy and lipid metabolism. J Biol Chem 287, 39107-39114 https://doi.org/10.1074/jbc.M112.412569
  35. Xu P, Das M, Reilly J and Davis RJ (2011) JNK regulates FoxO-dependent autophagy in neurons. Genes Dev 25, 310-322 https://doi.org/10.1101/gad.1984311
  36. Kume S, Uzu T, Horiike K et al (2010) Calorie restriction enhances cell adaptation to hypoxia through Sirt1-dependent mitochondrial autophagy in mouse aged kidney. J Clin Invest 120, 1043-1055 https://doi.org/10.1172/JCI41376
  37. Shinojima N, Yokoyama T, Kondo Y and Kondo S (2007) Roles of the Akt/mTOR/p70S6K and ERK1/2 signaling pathways in curcumin-induced autophagy. Autophagy 3, 635-637 https://doi.org/10.4161/auto.4916
  38. Divya MS, Roshin GE, Divya TS et al (2012) Umbilical cord blood-derived mesenchymal stem cells consist of a unique population of progenitors co-expressing mesenchymal stem cell and neuronal markers capable of instantaneous neuronal differentiation. Stem Cell Res Ther 3, 57 https://doi.org/10.1186/scrt148
  39. Reddy NP, Vemuri MC and Pallu R (2007) Isolation of stem cells from human umbilical cord blood. Methods Mol Biol 407, 149-163
  40. Wang M, Yang Y, Yang D et at (2009) The immunomodulatory activity of human umbilical cord bloodderived mesenchymal stem cells in vitro. Immunology 126, 220-232 https://doi.org/10.1111/j.1365-2567.2008.02891.x