DOI QR코드

DOI QR Code

Oocyte quality is closely linked to DRP1 derived-mitochondrial fission and mitophagy by the NAD+ biosynthesis in a postovulatory-aging model of pigs

  • Ji-Hyun Shin (Department of Biotechnology, Daegu University) ;
  • Seul-Gi Yang (DU Center for Infertility, Daegu University) ;
  • Hyo-Jin Park (Department of Biotechnology, Daegu University) ;
  • Deog-Bon Koo (Department of Biotechnology, Daegu University)
  • Received : 2024.03.18
  • Accepted : 2024.04.22
  • Published : 2024.06.30

Abstract

Background: Post-ovulatory aging (POA) of oocytes is related to a decrease in the quality and quantity of oocytes caused by aging. Previous studies on the characteristics of POA have investigated injury to early embryonic developmental ability, but no information is available on its effects on mitochondrial fission and mitophagy-related responses. In this study, we aimed to elucidate the molecular mechanisms underlying mitochondrial fission and mitophagy in in vitro maturation (IVM) oocytes and a POA model based on RNA sequencing analysis. Methods: The POA model was obtained through an additional 24 h culture following the IVM of matured oocytes. NMN treatment was administered at a concentration of 25 μM during the oocyte culture process. We conducted MitoTracker staining and Western blot experiments to confirm changes in mitochondrial function between the IVM and POA groups. Additionally, comparative transcriptome analysis was performed to identify differentially expressed genes and associated changes in mitochondrial dynamics between porcine IVM and POA model oocytes. Results: In total, 32 common genes of apoptosis and 42 mitochondrial fission and function uniquely expressed genes were detected (≥ 1.5-fold change) in POA and porcine metaphase II oocytes, respectively. Functional analyses of mitochondrial fission, oxidative stress, mitophagy, autophagy, and cellular apoptosis were observed as the major changes in regulated biological processes for oocyte quality and maturation ability compared with the POA model. Additionally, we revealed that the activation of NAD+ by nicotinamide mononucleotide not only partly improved oocyte quality but also mitochondrial fission and mitophagy activation in the POA porcine model. Conclusions: In summary, our data indicate that mitochondrial fission and function play roles in controlling oxidative stress, mitophagy, and apoptosis during maturation in POA porcine oocytes. Additionally, we found that NAD+ biosynthesis is an important pathway that mediates the effects of DRP1-derived mitochondrial morphology, dynamic balance, and mitophagy in the POA model.

Keywords

Acknowledgement

This research was supported by the National Research Foundation of Korea (NRF) funded by the Korean government (MSIT) (NRF-2021R1C1C2009469 and NRF2022R1A2C1002800), and the Basic Science Research Program through the NRF funded by the Ministry of Education (RS-2023-00246139), Republic of Korea.

References

  1. Agung B, Otoi T, Fuchimoto D, Senbon S, Onishi A, Nagai T. 2013. In vitro fertilization and development of porcine oocytes matured in follicular fluid. J. Reprod. Dev. 59:103-106.
  2. Appeltant R, Somfai T, Maes D, VAN Soom A, Kikuchi K. 2016. Porcine oocyte maturation in vitro: role of cAMP and oocyte-secreted factors - a practical approach. J. Reprod. Dev. 62:439-449.
  3. Bertoldo MJ, Listijono DR, Ho WJ, Riepsamen AH, Goss DM, Richani D, Jin XL, Mahbub S, Campbell JM, Habibalahi A, Loh WN, Youngson NA, Maniam J, Wong ASA, Selesniemi K, Bustamante S, Li C, Zhao Y, Marinova MB, Kim LJ, Lau L, Wu RM, Mikolaizak AS, Araki T, Le Couteur DG, Turner N, Morris MJ, Walters KA, Goldys E, O'Neill C, Gilchrist RB, Sinclair DA, Homer HA, Wu LE. 2020. NAD+ repletion rescues female fertility during reproductive aging. Cell Rep. 30:1670-1681. e7.
  4. Casillas F, Betancourt M, Cuello C, Ducolomb Y, Lopez A, Juarez-Rojas L, Retana-Marquez S. 2018. An efficiency comparison of different in vitro fertilization methods: IVF, ICSI, and PICSI for embryo development to the blastocyst stage from vitrified porcine immature oocytes. Porcine Health Manag. 4:16.
  5. Chen H, Detmer SA, Ewald AJ, Griffin EE, Fraser SE, Chan DC. 2003. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J. Cell Biol. 160:189-200.
  6. Chen RR, Wang J, Zhang M, Kong QQ, Sun GY, Jin CH, Luo MJ, Tan JH. 2022. Restraint stress of female mice during oocyte development facilitates oocyte postovulatory aging. Aging (Albany N.Y.) 14:9186-9199.
  7. Chen W, Zhao H, Li Y. 2023. Mitochondrial dynamics in health and disease: mechanisms and potential targets. Signal Transduct. Target. Ther. 8:333.
  8. Combelles CM, Gupta S, Agarwal A. 2009. Could oxidative stress influence the in-vitro maturation of oocytes? Reprod. Biomed. Online 18:864-880.
  9. Covarrubias AJ, Perrone R, Grozio A, Verdin E. 2021. NAD+ metabolism and its roles in cellular processes during ageing. Nat. Rev. Mol. Cell Biol. 22:119-141.
  10. Di Nisio V, Antonouli S, Damdimopoulou P, Salumets A, Cecconi S. 2022. In vivo and in vitro postovulatory aging: when time works against oocyte quality? J. Assist. Reprod. Genet. 39:905-918.
  11. Guo C, Sun L, Chen X, Zhang D. 2013. Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regen. Res. 8:2003-2014.
  12. Hall AR, Burke N, Dongworth RK, Hausenloy DJ. 2014. Mitochondrial fusion and fission proteins: novel therapeutic targets for combating cardiovascular disease. Br. J. Pharmacol. 171:1890-1906.
  13. Hong W, Mo F, Zhang Z, Huang M, Wei X. 2020. Nicotinamide mononucleotide: a promising molecule for therapy of diverse diseases by targeting NAD+ metabolism. Front. Cell Dev. Biol. 8:246.
  14. Hu B, Wang R, Wu D, Long R, Ruan J, Jin L, Ma D, Sun C, Liao S. 2023. Prospects for fertility preservation: the ovarian organ function reconstruction techniques for oogenesis, growth and maturation in vitro. Front. Physiol. 14:1177443.
  15. Iljas JD, Wei Z, Homer HA. 2020. Sirt1 sustains female fertility by slowing age-related decline in oocyte quality required for post-fertilization embryo development. Aging Cell 19:e13204.
  16. Jang SY, Kang HT, Hwang ES. 2012. Nicotinamide-induced mitophagy: event mediated by high NAD+/NADH ratio and SIRT1 protein activation. J. Biol. Chem. 287:19304-19314.
  17. Jiang Z and Shen H. 2022. Mitochondria: emerging therapeutic strategies for oocyte rescue. Reprod. Sci. 29:711-722.
  18. Kim JY, Zhou D, Cui XS. 2021. Bezafibrate prevents aging in in vitro-matured porcine oocytes. J. Anim. Sci. Technol. 63:766-777.
  19. Kim YH, Lee SY, Kim EY, Kim KH, Koong MK, Lee KA. 2022. The antioxidant auraptene improves aged oocyte quality and embryo development in mice. Antioxidants (Basel) 12:87.
  20. Klimova N, Long A, Kristian T. 2019a. Nicotinamide mononucleotide alters mitochondrial dynamics by SIRT3-dependent mechanism in male mice. J. Neurosci. Res. 97:975-990.
  21. Koroglu N and Aydin T. 2023. Oocyte vitrification for oncological and social reasons. Turk. J. Obstet. Gynecol. 20:59-63.
  22. Liang X and Dang S. 2021. Mitochondrial dynamics related genes-MFN1, MFN2 and DRP1 polymorphisms are associated with risk of lung cancer. Pharmgenomics. Pers. Med. 14:695-703.
  23. Liu L, Huang J, He A, Zhou S, Lu Q, Yu X, Bai X, Li M, Huo R. 2022. 4,4'-dimethoxychalcone increases resistance of mouse oocytes to postovulatory aging in vitro. Reprod. Biomed. Online 44:411-422.
  24. Liu YJ, McIntyre RL, Janssens GE, Houtkooper RH. 2020. Mitochondrial fission and fusion: a dynamic role in aging and potential target for age-related disease. Mech. Ageing Dev. 186:111212.
  25. Martin JH, Nixon B, Cafe SL, Aitken RJ, Bromfield EG, Lord T. 2022. Oxidative Stress and Reproductive Function: oxidative stress and in vitro ageing of the post-ovulatory oocyte: an update on recent advances in the field. Reproduction 164: F109-F124.
  26. Miao Y, Cui Z, Gao Q, Rui R, Xiong B. 2020. Nicotinamide mononucleotide supplementation reverses the declining quality of maternally aged oocytes. Cell Rep. 32:107987.
  27. Miao Y, Cui Z, Zhu X, Gao Q, Xiong B. 2022. Supplementation of nicotinamide mononucleotide improves the quality of postovulatory aged porcine oocytes. J. Mol. Cell Biol. 14: mjac025.
  28. Miao Y, Zhou C, Cui Z, Zhang M, ShiYang X, Lu Y, Xiong B. 2018. Postovulatory aging causes the deterioration of porcine oocytes via induction of oxidative stress. FASEB J. 32:1328-1337.
  29. Mihalas BP, Redgrove KA, McLaughlin EA, Nixon B. 2017. Molecular mechanisms responsible for increased vulnerability of the ageing oocyte to oxidative damage. Oxid. Med. Cell. Longev. 2017:4015874.
  30. Moghadam ARE, Moghadam MT, Hemadi M, Saki G. 2022. Oocyte quality and aging. JBRA Assist. Reprod. 26:105-122.
  31. Moreira OC, Estebanez B, Martinez-Florez S, de Paz JA, Cuevas MJ, Gonzalez-Gallego J. 2017. Mitochondrial function and mitophagy in the elderly: effects of exercise. Oxid. Med. Cell. Longev. 2017:2012798.
  32. Nagamatsu G. 2023. Oocyte aging in comparison to stem cells in mice. Front. Aging 4:1158510.
  33. Nicholas C, Darmon S, Patrizio P, Albertini DF, Barad DH, Gleicher N. 2023. Changing clinical significance of oocyte maturity grades with advancing female age advances precision medicine in IVF. iScience 26:107308.
  34. Pollard CL, Gibb Z, Hawdon A, Swegen A, Grupen CG. 2021. Supplementing media with NAD+ precursors enhances the in vitro maturation of porcine oocytes. J. Reprod. Dev. 67:319-326.
  35. Sasaki H, Hamatani T, Kamijo S, Iwai M, Kobanawa M, Ogawa S, Miyado K, Tanaka M. 2019. Impact of oxidative stress on age-associated decline in oocyte developmental competence. Front. Endocrinol. (Lausanne) 10:811.
  36. Song M, Li Y, Zhou Y, Yan J, Zhou X, Gao Q, Miao Y, Xiong B. 2022. Nicotinamide mononucleotide supplementation improves the quality of porcine oocytes under heat stress. J. Anim. Sci. Biotechnol. 13:68.
  37. Srivastava S. 2017. The mitochondrial basis of aging and agerelated disorders. Genes (Basel) 8:398.
  38. Sun YL, Tang SB, Shen W, Yin S, Sun QY. 2019a. Roles of Resveratrol in improving the quality of postovulatory aging oocytes in vitro. Cells 8:1132.
  39. Wang Y, Dai X, Li H, Jiang H, Zhou J, Zhang S, Guo J, Shen L, Yang H, Lin J, Yan H. 2023. The role of mitochondrial dynamics in disease. MedComm (2020) 4:e462.
  40. Wen X, Yang Q, Sun D, Jiang ZY, Wang T, Liu HR, Han Z, Wang L, Liang CG. 2023. Cumulus cells accelerate postovulatory oocyte aging through IL1-IL1R1 interaction in mice. Int. J. Mol. Sci. 24:3530.
  41. Wilcox AJ, Weinberg CR, Baird DD. 1998. Post-ovulatory ageing of the human oocyte and embryo failure. Hum. Reprod. 13: 394-397.
  42. Xing X, Liang Y, Li Y, Zhao Y, Zhang Y, Li Z, Li Z, Wu Z. 2023. Fisetin delays postovulatory oocyte aging by regulating oxidative stress and mitochondrial function through Sirt1 pathway. Molecules 28:5533.
  43. Yang Q, Zhu L, Jin L. 2020. Human follicle in vitro culture including activation, growth, and maturation: a review of research progress. Front. Endocrinol. (Lausanne) 11:548.
  44. Yang SG, Park HJ, Kim JW, Jung JM, Kim MJ, Jegal HG, Kim IS, Kang MJ, Wee G, Yang HY, Lee YH, Seo JH, Kim SU, Koo DB. 2018. Mito-TEMPO improves development competence by reducing superoxide in preimplantation porcine embryos. Sci. Rep. 8:10130.
  45. Ye C, Chen P, Xu B, Jin Y, Pan Y, Wu T, Du Y, Mao J, Wu R. 2023. Abnormal expression of fission and fusion genes and the morphology of mitochondria in eutopic and ectopic endometrium. Eur. J. Med. Res. 28:209.
  46. Zerihun M, Sukumaran S, Qvit N. 2023. The Drp1-mediated mitochondrial fission protein interactome as an emerging core player in mitochondrial dynamics and cardiovascular disease therapy. Int. J. Mol. Sci. 24:5785.
  47. Zhao ZH, Meng TG, Li A, Schatten H, Wang ZB, Sun QY. 2020. RNA-Seq transcriptome reveals different molecular responses during human and mouse oocyte maturation and fertilization. BMC Genomics 21:475.