DOI QR코드

DOI QR Code

Vitrification, in vitro fertilization, and development of Atg7 deficient mouse oocytes

  • Bang, Soyoung (Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University) ;
  • Lee, Geun-Kyung (Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University) ;
  • Shin, Hyejin (Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University) ;
  • Suh, Chang Suk (Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital) ;
  • Lim, Hyunjung Jade (Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University)
  • Received : 2016.01.11
  • Accepted : 2016.02.10
  • Published : 2016.03.31

Abstract

Objective: Autophagy contributes to the clearance and recycling of macromolecules and organelles in response to stress. We previously reported that vitrified mouse oocytes show acute increases in autophagy during warming. Herein, we investigate the potential role of Atg7 in oocyte vitrification by using an oocyte-specific deletion model of the Atg7 gene, a crucial upstream gene in the autophagic pathway. Methods: Oocyte-specific Atg7 deficient mice were generated by crossing Atg7 floxed mice and Zp3-Cre transgenic mice. The oocytes were vitrified-warmed and then subjected to in vitro fertilization and development. The rates of survival, fertilization, and development were assessed in the Atg7 deficient oocytes in comparison with the wildtype oocytes. Light chain 3 (LC3) immunofluorescence staining was performed to determine whether this method effectively evaluates the autophagy status of oocytes. Results: The survival rate of vitrified-warmed $Atg7^{f/f}$;Zp3-Cre ($Atg7^{d/d}$) metaphase II (MII) oocytes was not significantly different from that of the wildtype ($Atg7^{f/f}$) oocytes. Fertilization and development in the $Atg7^{d/d}$ oocytes were significantly lower than the $Atg7^{f/f}$ oocytes, comparable to the $Atg5^{d/d}$ oocytes previously described. Notably, the developmental rate improved slightly in vitrified-warmed $Atg7^{d/d}$ MII oocytes when compared to fresh $Atg7^{d/d}$ oocytes. LC3 immunofluorescence staining showed that this method can be reliably used to assess autophagic activation in oocytes. Conclusion: We confirmed that the LC3-positive signal is nearly absent in $Atg7^{d/d}$ oocytes. While autophagy is induced during the warming process after vitrification of MII oocytes, the Atg7 gene is not essential for survival of vitrified-warmed oocytes. Thus, induction of autophagy during warming of vitrified MII oocytes seems to be a natural response to manage cold or other cellular stresses.

Keywords

References

  1. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 2012;8:445-544. https://doi.org/10.4161/auto.19496
  2. Boya P, Reggiori F, Codogno P. Emerging regulation and functions of autophagy. Nat Cell Biol 2013;15:713-20. https://doi.org/10.1038/ncb2788
  3. Yang Z, Klionsky DJ. Eaten alive: a history of macroautophagy. Nat Cell Biol 2010;12:814-22. https://doi.org/10.1038/ncb0910-814
  4. Choi S, Shin H, Song H, Lim HJ. Suppression of autophagic activation in the mouse uterus by estrogen and progesterone. J Endocrinol 2014;221:39-50. https://doi.org/10.1530/JOE-13-0449
  5. Ohsumi Y. Molecular dissection of autophagy: two ubiquitin-like systems. Nat Rev Mol Cell Biol 2001;2:211-6. https://doi.org/10.1038/35056522
  6. Ichimura Y, Kirisako T, Takao T, Satomi Y, Shimonishi Y, Ishihara N, et al. A ubiquitin-like system mediates protein lipidation. Nature 2000;408:488-92. https://doi.org/10.1038/35044114
  7. Komatsu M, Waguri S, Ueno T, Iwata J, Murata S, Tanida I, et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol 2005;169:425-34. https://doi.org/10.1083/jcb.200412022
  8. Hale AN, Ledbetter DJ, Gawriluk TR, Rucker EB 3rd. Autophagy: regulation and role in development. Autophagy 2013;9:951-72. https://doi.org/10.4161/auto.24273
  9. Epifano O, Liang LF, Familari M, Moos MC Jr, Dean J. Coordinate expression of the three zona pellucida genes during mouse oogenesis. Development 1995;121:1947-56.
  10. de Vries WN, Binns LT, Fancher KS, Dean J, Moore R, Kemler R, et al. Expression of Cre recombinase in mouse oocytes: a means to study maternal effect genes. Genesis 2000;26:110-2. https://doi.org/10.1002/(SICI)1526-968X(200002)26:2<110::AID-GENE2>3.0.CO;2-8
  11. Bang S, Shin H, Song H, Suh CS, Lim HJ. Autophagic activation in vitrified-warmed mouse oocytes. Reproduction 2014;148:11-9. https://doi.org/10.1530/REP-14-0036
  12. Martinez-Burgos M, Herrero L, Megias D, Salvanes R, Montoya MC, Cobo AC, et al. Vitrification versus slow freezing of oocytes:effects on morphologic appearance, meiotic spindle configuration, and DNA damage. Fertil Steril 2011;95:374-7. https://doi.org/10.1016/j.fertnstert.2010.07.1089
  13. Gualtieri R, Mollo V, Barbato V, Fiorentino I, Iaccarino M, Talevi R. Ultrastructure and intracellular calcium response during activation in vitrified and slow-frozen human oocytes. Hum Reprod 2011;26:2452-60. https://doi.org/10.1093/humrep/der210
  14. Lim HJ, Song H. Evolving tales of autophagy in early reproductive events. Int J Dev Biol 2014;58:183-7. https://doi.org/10.1387/ijdb.130337hl
  15. Practice Committees of American Society for Reproductive Medicine; Society for Assisted Reproductive Technology. Mature oocyte cryopreservation: a guideline. Fertil Steril 2013;99:37-43. https://doi.org/10.1016/j.fertnstert.2012.09.028
  16. Cha SK, Kim BY, Kim MK, Kim YS, Lee WS, Yoon TK, et al. Effects of various combinations of cryoprotectants and cooling speed on the survival and further development of mouse oocytes after vitrification. Clin Exp Reprod Med 2011;38:24-30. https://doi.org/10.5653/cerm.2011.38.1.24
  17. Tsukamoto S, Kuma A, Murakami M, Kishi C, Yamamoto A, Mizushima N. Autophagy is essential for preimplantation development of mouse embryos. Science 2008;321:117-20. https://doi.org/10.1126/science.1154822
  18. Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell 2004;15:1101-11. https://doi.org/10.1091/mbc.e03-09-0704
  19. Kuma A, Matsui M, Mizushima N. LC3, an autophagosome marker, can be incorporated into protein aggregates independent of autophagy: caution in the interpretation of LC3 localization. Autophagy 2007;3:323-8. https://doi.org/10.4161/auto.4012
  20. Lee GK, Shin H, Lim HJ. Rapamycin influences the efficiency of in vitro fertilization and development in the mouse: a role for autophagic activation. Asian-Australas J Anim Sci 2015 Oct 8 [Epub]. http://dx.doi.org/10.5713/ajas.15.0762.

Cited by

  1. Autocrine actions of prolactin contribute to the regulation of lactotroph function in vivo vol.32, pp.9, 2016, https://doi.org/10.1096/fj.201701111rr
  2. Contextualizing Autophagy during Gametogenesis and Preimplantation Embryonic Development vol.22, pp.12, 2021, https://doi.org/10.3390/ijms22126313