DOI QR코드

DOI QR Code

Effects of Mito-TEMPO on the survival of vitrified bovine blastocysts in vitro

  • Jeong, Jae-Hoon (Department of Biotechnology, College of Engineering, Daegu University) ;
  • Yang, Seul-Gi (Department of Biotechnology, College of Engineering, Daegu University) ;
  • Park, Hyo-Jin (Department of Biotechnology, College of Engineering, Daegu University) ;
  • Koo, Deog-Bon (Department of Biotechnology, College of Engineering, Daegu University)
  • Received : 2021.11.23
  • Accepted : 2021.11.30
  • Published : 2021.12.31

Abstract

Vitrification methods are commonly used for mammalian reproduction through the long-term storage of blastocyst produced in vitro. However, the survival and quality of embryos following vitrification are significantly low compared with blastocyst from in vitro production (IVP). This study evaluates that the survival of frozen-thawed bovine embryos was relevant to mitochondrial superoxide derived mitochondrial activity. Here we present supplementation of the cryopreservation medium with Mito-TEMPO (0.1 µM) induced a significant (p < 0.001; non-treated group: 56.8 ± 8.7%, reexpanded at 24 h vs Mito-TEMPO treated group: 77.5 ± 8.9%, re-expanded at 24 h) improvement in survival rate of cryopreserved-thawed bovine blastocyst. To confirm the quality of vitrified blastocyst after thawing, DNA fragmentation of survived embryos was examined by TUNEL assay. As a result, TUNEL positive cells rates of frozen-thawed embryos were lower in the Mito-TEMPO treated group (4.2 ± 1.4%) than the non-treated group (7.1 ± 3.5%). In addition, we investigated the intracellular ROS and mitochondrial specific superoxide production using DCF-DA and Mito-SOX staining in survived bovine embryos following vitrification depending on Mito-TEMPO treatment. As expected, intracellular ROS levels and superoxide production of vitrified blastocysts after cryopreservation were significantly reduced (p < 0.05) according to Mito-TEMPO supplement in freezing medium. Also, mitochondrial activity measured by MitoTracker Orange staining increased in the frozen-thawed embryos with Mito-TEMPO compared with non-treated group. These results indicate that the treatment of Mito-TEMPO during cryopreservation might induce reduction in DNA fragmentation and apoptosis-related ROS production, consequently increasing mitochondrial activation for developmental capacity of frozen-thawed embryos.

Keywords

Acknowledgement

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF-2021R1C1C2009469 and NRF2019R1A2C1085199) funded by and the Ministry of Science and ICT, Republic of Korea.

References

  1. Al-Mutary MG. 2021. Use of antioxidants to augment semen efficiency during liquid storage and cryopreservation in livestock animals: a review. J. King Saud Univ. Sci. 33:101226. https://doi.org/10.1016/j.jksus.2020.10.023
  2. Asadzadeh N, Abdollahi Z, Masoudi R. 2021. Fertility and flow cytometry evaluations of ram frozen semen in plant-based extender supplemented with Mito-TEMPO. Anim. Reprod. Sci. 233:106836. https://doi.org/10.1016/j.anireprosci.2021.106836
  3. Babayev E and Seli E. 2015. Oocyte mitochondrial function and reproduction. Curr. Opin. Obstet. Gynecol. 27:175-181. https://doi.org/10.1097/GCO.0000000000000164
  4. Chang H, Chen H, Zhang L, Wang Y, Xie X, Quan F. 2019. Effect of oocyte vitrification on DNA damage in metaphase II oocytes and the resulting preimplantation embryos. Mol. Reprod. Dev. 86:1603-1614. https://doi.org/10.1002/mrd.23247
  5. Choumar A, Tarhuni A, Letteron P, Reyl-Desmars F, Dauhoo N, Damasse J, Vadrot N, Nahon P, Moreau R, Mansouri A. 2011. Lipopolysaccharide-induced mitochondrial DNA depletion. Antioxid. Redox Signal. 15:2837-2854. https://doi.org/10.1089/ars.2010.3713
  6. Dikalova AE, Bikineyeva AT, Budzyn K, Nazarewicz RR, McCann L, Lewis W, Dikalov SI. 2010. Therapeutic targeting of mitochondrial superoxide in hypertension. Circ. Res. 107:106-116. https://doi.org/10.1161/CIRCRESAHA.109.214601
  7. Elahi F, Shin H, Lee E. 2017. Endoplasmic stress inhibition during oocyte maturation improves preimplantation development of cloned pig embryos. J. Emb. Trans. 32:287-295. https://doi.org/10.12750/JET.2017.32.4.287
  8. Gualtieri R, Kalthur G, Barbato V, Di Nardo M, Talevi R. 2021. Mitochondrial dysfunction and oxidative stress caused by cryopreservation in reproductive cells. Antioxidants (Basel) 10:337. https://doi.org/10.3390/antiox10030337
  9. Guo C, Sun L, Zhang D. 2013. Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regen. Res. 8:2003-2014.
  10. Gupta MK and Lee HT. 2010. Cryopreservation of oocytes and embryos by vitrification. Korean J. Reprod. Med. 37:267-291.
  11. Hara T, Kin A, Aoki S, Nakamura S, Shirasuna K, Iwata H. 2018. Resveratrol enhances the clearance of mitochondrial damage by vitrification and improves the development of vitrified-warmed bovine embryos. PLoS One 13:e0204571. https://doi.org/10.1371/journal.pone.0204571
  12. Huebinger J, Han HM, Hofnagel O, Vetter IR, Grabenbauer M. 2016. Direct measurement of water states in cryopreserved cells reveals tolerance toward ice crystallization. Biophys. J. 110:840-849. https://doi.org/10.1016/j.bpj.2015.09.029
  13. Inaba Y, Miyashita S, Somfai T, Geshi M, Matoba S, Nagai T. 2016. Cryopreservation method affects DNA fragmentation in trophectoderm and the speed of re-expansion in bovine blastocysts. Cryobiology 72:86-92. https://doi.org/10.1016/j.cryobiol.2016.03.006
  14. Kim JW, Yang SG, Park HJ, Kim JH, Lee DM, Woo SM, Kim HJ, Kim HA, Jeong JH, Koo DB. 2020. Comparison of Cryotop and ReproCarreir products for cryopreservation of bovine blastocysts through survival rate and blastocysts quality. J. Anim. Reprod. Biotechnol. 35:207-213. https://doi.org/10.12750/JARB.35.2.207
  15. Lee HL, Kim YJ. 2014. Studies on cryotop vitrification method for simple freezing of Hanwoo embryos. J. Emb. Trans. 29:13-19. https://doi.org/10.12750/JET.2014.29.1.13
  16. Lee KS, Kim EY, Jeon K, Cho SG, Han YJ, Yang BC, Lee SS, Ko MS, Riu KJ, Park SP. 2011. 3,4-Dihydroxyflavone acts as an antioxidant and antiapoptotic agent to support bovine embryo development in vitro. J. Reprod. Dev. 57:127-134. https://doi.org/10.1262/jrd.10-029A
  17. Lee S, Park HW, Cheong HT, Yang BK. 2016. Effects of turine and vitamin E on sperm viability, membrane integrity and mitochondrial activity damaged by bromopropane in fresh boar semen. J. Emb. Trans. 31:13-17. https://doi.org/10.12750/JET.2016.31.1.13
  18. Len JS, Tan SX. 2019. The roles of reactive oxygen species and antioxidants in cryopreservation. Biosci. Rep. 39:BSR20191601. https://doi.org/10.1042/bsr20191601
  19. Lu X, Zhang Y, Bai H, Liu J, Wu B. 2018. Mitochondria-targeted antioxidant MitoTEMPO improves the post-thaw sperm quality. Cryobiology 80:26-29. https://doi.org/10.1016/j.cryobiol.2017.12.009
  20. Majidi Gharenaz N, Movahedin M, Pour Beiranvand S. 2016. Alternation of apoptotic and implanting genes expression of mouse embryos after re-vitrification. Int. J. Reprod. Biomed. 14:511-518. https://doi.org/10.29252/ijrm.14.8.511
  21. Marsico TV, de Camargo J, Sudano MJ. 2019. Embryo competence and cryosurvival: molecular and cellular features. Anim. Reprod. 16:423-439. https://doi.org/10.21451/1984-3143-AR2019-0072
  22. Masoudi R, Sharafi M. 2021. Effects of freezing extender supplementation with mitochondria-targeted antioxidant Mito-TEMPO on frozen-thawed rooster semen quality and reproductive performance. Anim. Reprod. Sci. 225:106671. https://doi.org/10.1016/j.anireprosci.2020.106671
  23. May-Panloup P, Boguenet M, Hachem HE, Reynier P. 2021. Embryo and its mitochondria. Antioxidants (Basel) 10:139. https://doi.org/10.3390/antiox10020139
  24. Nohales-Corcoles M, Sevillano-Almerich G, Di Emidio G, Tatone C, Cobo AC, De Los Santos Molina MJ. 2016. Impact of vitrification on the mitochondrial activity and redox homeostasis of human oocyte. Hum. Reprod. 31:1850-1858. https://doi.org/10.1093/humrep/dew130
  25. Ott M, Gogvadze V, Zhivotovsky B. 2007. Mitochondria, oxidative stress and cell death. Apoptosis 12:913-922. https://doi.org/10.1007/s10495-007-0756-2
  26. Park HJ, Song BS, Kim JW, Yang SG, Koo DB. 2020. Exposure of triclosan in porcine oocyte leads to superoxide production and mitochondrial-mediated apoptosis during in vitro maturation. Int. J. Mol. Sci. 21:3050. https://doi.org/10.3390/ijms21093050
  27. Shadel GS and Horvath TL. 2015. Mitochondrial ROS signaling in organismal homeostasis. Cell 163:560-569. https://doi.org/10.1016/j.cell.2015.10.001
  28. Stoecklein KS, Ortega MS, Spate LD, Prather RS. 2021. Improved cryopreservation of in vitro produced bovine embryos using FGF2, LIF, and IGF1. PLoS One 16:e0243727. https://doi.org/10.1371/journal.pone.0243727
  29. van Hameren G, Campbell G, Deck M, Berthelot J, Gautier B, Quintana P, Tricaud N. 2019. In vivo real-time dynamics of ATP and ROS production in axonal mitochondria show decoupling in mouse models of peripheral neuropathies. Acta Neuropathol. Commun. 7:86. https://doi.org/10.1186/s40478-019-0740-4
  30. Xia M, Zhang Y, Jin K, Lu Z, Xiong W. 2019. Communication between mitochondria and other organelles: a brand-new perspective on mitochondria in cancer. Cell Biosci. 9:27. https://doi.org/10.1186/s13578-019-0289-8
  31. Yang SG, Park HJ, Kim JW, Jung JM, Kim MJ, Jegal HG, Kim IS, Kang MJ, Wee G, Yang HY, Lee YH, Seo JH, Koo DB. 2018. Mito-TEMPO improves development competence by reducing superoxide in preimplantation porcine embryos. Sci. Rep. 8:10130. https://doi.org/10.1038/s41598-018-28497-5
  32. Yang SG, Park HJ, Lee SM, Kim JW, Kim MJ, Kim IS, Koo DB. 2019. Reduction of mitochondrial derived superoxide by Mito-TEMPO improves porcine oocyte maturation in vitro. J. Anim. Reprod. Biotechnol. 34:10-19. https://doi.org/10.12750/JARB.34.1.10
  33. Yousefian I, Zare-Shahneh A, Goodarzi A, Fouladi-Nashta AA. 2021. The effect of Tempo and MitoTEMPO on oocyte maturation and subsequent embryo development in bovine model. Theriogenology 176:128-136. https://doi.org/10.1016/j.theriogenology.2021.09.016