DOI QR코드

DOI QR Code

Exogenous Nitric Oxide Donation During In Vitro Maturation Improves Embryonic Development after Parthenogenesis and Somatic Cell Nuclear Transfer in Pigs

  • Elahi, Fazle (College of Veterinary Medicine, Kangwon National University) ;
  • Shin, Hyeji (College of Veterinary Medicine, Kangwon National University) ;
  • Lee, Joohyeong (Institute of Veterinary Science, Kangwon National University) ;
  • Lee, Seung Tae (Division of Applied Animal Science, College of Animal Life Science, Kangwon National University) ;
  • Lee, Geun-Shik (College of Veterinary Medicine, Kangwon National University) ;
  • Lee, Eunsong (College of Veterinary Medicine, Kangwon National University)
  • Received : 2018.11.23
  • Accepted : 2018.12.13
  • Published : 2018.12.31

Abstract

Nitric oxide (NO) has an important role in oocyte maturation and embryonic development in mammals. This study examined the effect of exogenous NO donor S-nitroso-N-acetylpenicillamine (SNAP) in a maturation medium on meiotic progression and embryonic development after parthenogenesis (PA) and somatic cell nuclear transfer (SCNT) in pigs. When oocytes were exposed to $0.1{\mu}M$ SNAP for first 22 h of in vitro maturation (IVM) in Experiment 1, SNAP significantly improved blastocyst development in both defined and standard follicular fluid-supplemented media compared to untreated control (48.4 vs. 31.7-42.5%). SNAP treatment significantly arrested meiotic progression of oocytes at the germinal vesicle stage at 11 h of IVM (61.2 vs. 38.7%). However, there was no effect on meiotic progression at 22 h of IVM (Experiment 2). In Experiment 3, when oocytes were treated with SNAP at 0.001, 0.1 and $10{\mu}M$ during the first 22 h of IVM to determine a suitable concentration, $0.1{\mu}M$ SNAP (54.2%) exhibited a higher blastocyst formation than 0 and $10{\mu}M$ SNAP (36.6 and 36.6%, respectively). Time-dependent effect of SNAP treatment was evaluated in Experiment 4. It was observed that SNAP treatment for the first 22 h of IVM significantly increased blastocyst formation compared to no treatment (57.1% vs. 46.2%). Antioxidant effect of SNAP was compared with that of cysteine. SNAP treatment significantly improved embryonic development to the blastocyst stage (49.1-51.5% vs. 34.4-37.5%) irrespective of the presence or absence of cysteine (Experiment 5). Moreover, SNAP significantly increased glutathione (GSH) content and inversely decreased the reactive oxygen species (ROS) level and mitochondrial oxidative activity in IVM oocytes. SNAP treatment during IVM showed a stimulating effect on in vitro development of SCNT embryos (Experiment 7). These results demonstrates that SNAP improves developmental competence of PA and SCNT embryos probably by maintaining the redox homeostasis through increasing GSH content and mitochondrial quality and decreasing ROS in IVM oocytes.

Keywords

References

  1. Abbasi M, Akbari M, Amidi F, RagerdiKashani I, Mahmoudi R, Sobhani A, Takzare N, Pasbakhsh P, BarbarestaniM, Abolhassani F, Sato E. 2009. Nitric oxide acts through different signalling pathways in maturation of cumulus cell-enclosed mouse oocytes. DARU J. Pharm. Sci. 17: 8-52.
  2. Abe K, Matsuoka K, Inoue N, Taga M, Kato T. 1999. Messenger RNA of neuronal nitric oxide synthase is expressed and possibly functions in mouse oocytes and embryos during preimplantation development. Biomedical Research 20: 61-65. https://doi.org/10.2220/biomedres.20.61
  3. Barroso RP, Osuamkpe C, Nagamani M, Yallampalli C. 1998. Nitric oxide inhibits development of embryos and implantation in mice. Mol. Hum. Reprod. 4: 503-507. https://doi.org/10.1093/molehr/4.5.503
  4. Basini G, Bianchi F, Bussolati S, Baioni L, Ramoni R, Grolli S, Conti V, Bianchi F, Grasselli F. 2012. Atrazine disrupts steroidogenesis, VEGF and NO production in swine granulosa cells. Ecotoxicol. Environ. Saf. 85: 59-63. https://doi.org/10.1016/j.ecoenv.2012.08.027
  5. Bergandi L, Basso G, Evangelista F, Canosa S, Dalmasso P, Aldieri E, Revelli A, Benedetto C, Ghigo D. 2014. Inducible nitric oxide synthase and heme oxygenase 1 are expressed in human cumulus cells and may be used as biomarkers of oocyte competence. Reprod. Sci. 21: 1370-1377. https://doi.org/10.1177/1933719114525268
  6. Bilodeau-Goeseels S. 2007. Effects of manipulating the nitric oxide/cyclic GMP pathway on bovine oocyte meiotic resumption in vitro. Theriogenology 68: 693-701. https://doi.org/10.1016/j.theriogenology.2007.05.063
  7. Bu S, Xia G, Tao Y, Lei L, Zhou B. 2003. Dual effects of nitric oxide on meiotic maturation of mouse cumulus cell-enclosed oocytes in vitro. Mol. Cell. Endocrinol. 207: 21-30. https://doi.org/10.1016/S0303-7207(03)00213-2
  8. Bu S, Xie H, Tao Y, Wang J, Xia G. 2004. Nitric oxide influences the maturation of cumulus cell-enclosed mouse oocytes cultured in spontaneous maturation medium and hypoxanthine-supplemented medium through different signaling pathways. Mol. Cell. Endocrinol. 223: 85-93. https://doi.org/10.1016/j.mce.2004.04.015
  9. Chmelikova E, Sedmikova M, Petr J, Kott T, Lanska V, Tumova L, Tichovska H, Jeseta M. 2009. Expression and localization of nitric oxide synthase isoforms during porcine oocyte growth and acquisition of meiotic competence. Czech. J. Anim. Sci. 54:137-149. https://doi.org/10.17221/1742-CJAS
  10. Curnow EC, Ryan JP, Saunders DM, Hayes ES. 2010. Developmental potential of bovine oocytes following IVM in the presence of glutathione ethyl ester. Reprod. Fertil. Dev. 22: 597-605. https://doi.org/10.1071/RD09228
  11. Fujitani Y, Kasai K, Ohtani S, Nishimura K, Yamada M, Utsumi K. 1997. Effect of oxygen concentration and free radicals on in vitro development of in vitro produced bovine embryos. J. Anim. Sci. 75: 483-489. https://doi.org/10.2527/1997.752483x
  12. Furnus CC, de Matos DG, Picco S, Garcia PP, Inda AM, Mattioli G, Errecalde AL. 2008. Metabolic requirements associated with GSH synthesis during in vitro maturation of cattle oocytes. Anim. Reprod. Sci. 109: 88-99. https://doi.org/10.1016/j.anireprosci.2007.12.003
  13. Goud PT, Goud AP, Najafi T, Gonik B, Diamond MP, Saed GM, Zhang X, Abu-Soud HM. 2014. Direct real-time measurement of intra-oocyte nitric oxide concentration in vivo. PLoS One 9: e98720. https://doi.org/10.1371/journal.pone.0098720
  14. Hess DT, Matsumoto A, Kim SO, Marshall HE, Stamler JS. 2005. Protein S-nitrosylation: purview and parameters. Nat. Rev. Mol. Cell. Biol. 6: 150-166. https://doi.org/10.1038/nrm1569
  15. Hunter RH, Polge C. 1966. Maturation of follicular oocytes in the pig after injection of human chorionic gonadotrophin. J. Reprod. Fertil. 12: 525-531. https://doi.org/10.1530/jrf.0.0120525
  16. Jablonka-Shariff A, Olson LM. 1998. The role of nitric oxide in oocyte meiotic maturation and ovulation: meiotic abnormalities of endothelial nitric oxide synthase knock-out mouse oocytes. Endocrinology 139: 2944-2954. https://doi.org/10.1210/endo.139.6.6054
  17. Jablonka-Shariff A, Olson LM. 2000. Nitric oxide is essential for optimal meiotic maturation of murine cumulus-oocyte complexes in vitro. Mol. Reprod. Dev. 55: 412-421. https://doi.org/10.1002/(SICI)1098-2795(200004)55:4<412::AID-MRD9>3.0.CO;2-W
  18. Jaffrey SR, Erdjument-Bromage H, Ferris CD, Tempst P, Snyder SH. 2001. Protein S-nitrosylation: a physiological signal for neuronal nitric oxide. Nat. Cell Biol. 3: 193-197. https://doi.org/10.1038/35055104
  19. Ji Y, Akerboom TP, Sies H, Thomas JA.1999. S-nitrosylation and S-glutathiolation of protein sulfhydryls by S-nitroso glutathione. Arch. Biochem. Biophys. 362: 67-78. https://doi.org/10.1006/abbi.1998.1013
  20. Katia RLS, Pedro RLP, Ligia GM, Marcos RC, Cláudia LVL. 2014. Effect of nitric oxide on the cyclic guanosine monophosphate (cGMP) pathway during meiosis resumption in bovine oocytes. Theriogenology, 81: 556-564 https://doi.org/10.1016/j.theriogenology.2013.11.008
  21. Koo OJ, Jang G, Kwon DK, Kang JT, Kwon OS, Park HJ, Kang SK, Lee BC. 2008. Electrical activation induces reactive oxygen species in porcine embryos. Theriogenology 70: 1111-1118. https://doi.org/10.1016/j.theriogenology.2008.06.031
  22. Kuo RC, Baxter GT, Thompson SH, Stricker SA, Patton C, Bonaventura J, Epel D. 2000. NO is necessary and sufficient for egg activation at fertilization. Nature 406: 633-636. https://doi.org/10.1038/35020577
  23. Kuo PC, Abe KY, Schroeder RA. 1996. Interleukin-1-induced nitric oxide production modulates glutathione synthesis in cultured rat hepatocytes. Am. J. Physiol., 271: 851-862. https://doi.org/10.1152/ajpcell.1996.271.3.C851
  24. Lee J, You J, Lee GS, Hyun SH, Lee E. 2013a. Pig oocytes with a large perivitelline space matured in vitro show greater developmental competence after parthenogenesis and somatic cell nuclear transfer. Mol. Reprod. Dev. 80:753-762.
  25. Lee TH, Lee MS, Huang CC, Tsao HM, Lin PM, Ho HN, Shew JY, Yang YS. 2013b. Nitric oxide modulates mitochondrial activity and apoptosis through protein S-nitrosylation for preimplantation embryo development. Journal of assisted reproduction and genetics 30: 1063-1072. https://doi.org/10.1007/s10815-013-0045-7
  26. Lee Y, Lee H, Park B, Elahi F, Lee J, Lee ST, Park CK, Hyun SH, Lee E. 2016. Alpha-linolenic acid treatment during oocyte maturation enhances embryonic development by influencing mitogen-activated protein kinase activity and intraoocyte glutathione content in pigs. J. Anim. Sci. 94: 3255-3263. https://doi.org/10.2527/jas.2016-0384
  27. Li Q, Miao DQ, Zhou P, Wu YG, Gao D, Wei DL, Cui W, Tan JH. 2011. Glucose metabolism in mouse cumulus cells prevents oocyte aging by maintaining both energy supply and the intracellular redox potential. Biol. Reprod. 84: 1111-1118. https://doi.org/10.1095/biolreprod.110.089557
  28. Luberda Z. 2005. The role of glutathione in mammalian gametes. Reprod. Biol. 5: 5-17.
  29. Matta SG, Caldas-Bussiere MC, Viana KS, Faes MR, Paes de Carvalho CS, Dias BL, Quirino CR. 2009. Effect of inhibition of synthesis of inducible nitric oxide synthase-derived nitric oxide by aminoguanidine on the in vitro maturation of oocytecumulus complexes of cattle. Anim. Reprod. Sci. 111: 189-201. https://doi.org/10.1016/j.anireprosci.2008.03.002
  30. Mizutani E, Ohta H, Kishigami S, Van Thuan N, Hikichi T, Wakayama S, Kosaka M, Sato E, Wakayama T. 2006. Developmental ability of cloned embryos from neural stem cells. Reproduction 132: 849-857. https://doi.org/10.1530/rep.1.01010
  31. Moawad AR, Xu B, Tan SL, Taketo T. 2014. L-carnitine supplementation during vitrification of mouse germinal vesicle stage-oocytes and their subsequent in vitro maturation improves meiotic spindle configuration and mitochondrial distribution in metaphase II oocytes. Hum. Reprod. 29: 2256-2268. https://doi.org/10.1093/humrep/deu201
  32. Nakamura Y, Yamagata Y, Sugino N, Takayama H. Kato H. 2002. Nitric oxide inhibits oocyte meiotic maturation. Biol. Reprod. 67: 1588-1592. https://doi.org/10.1095/biolreprod.102.005264
  33. Pires PR, Santos NP, Adona PR, Natori MM, Schwarz KR, de Bem TH, Leal CL. 2009. Endothelial and inducible nitric oxide synthases in oocytes of cattle. Animal reproduction science 116: 233-243. https://doi.org/10.1016/j.anireprosci.2009.02.019
  34. Sakatani M, Suda I, Oki T, Kobayashi S, Kobayashi S, Takahashi M. 2007. Effects of purple sweet potato anthocyanins on development and intracellular redox status of bovine preimplantation embryos exposed to heat shock. J. Reprod. Dev. 53: 605-614. https://doi.org/10.1262/jrd.18124
  35. Schwarz KR, Pires PR, Adona PR, Camara de Bem TH, Leal CL. 2008. Influence of nitric oxide during maturation on bovine oocyte meiosis and embryo development in vitro. Reprod. Fertil. Dev. 20: 529-536. https://doi.org/10.1071/RD07209
  36. Schwarz KR, Pires PR, Mesquita LG, Chiaratti MR, Leal CL. 2014. Effect of nitric oxide on the cyclic guanosine monophosphate (cGMP) pathway during meiosis resumption in bovine oocytes. Theriogenology 81: 556-564. https://doi.org/10.1016/j.theriogenology.2013.11.008
  37. Sela-Abramovich S, Edry I, Galiani D, Nevo N, Dekel N. 2006. Disruption of gap junctional communication within the ovarian follicle induces oocyte maturation. Endocrinology 147: 2280-2286. https://doi.org/10.1210/en.2005-1011
  38. Sengoku K, Takuma N, Horikawa M, Tsuchiya K, Komori H, Sharifa D, Tamate K, Ishikawa M. 2001. Requirement of nitric oxide for murine oocyte maturation, embryo development, and trophoblast outgrowth in vitro. Molecular reproduction and development 58: 262-268. https://doi.org/10.1002/1098-2795(200103)58:3<262::AID-MRD3>3.0.CO;2-8
  39. Somfai T, Ozawa M, Noguchi J, Kaneko H, Kuriani Karja NW, Farhudin M, Dinnyes A, Nagai T, Kikuchi K. 2007. Developmental competence of in vitro-fertilized porcine oocytes after in vitro maturation and solid surface vitrification: effect of cryopreservation on oocyte antioxidative system and cell cycle stage. Cryobiology 55: 115-126. https://doi.org/10.1016/j.cryobiol.2007.06.008
  40. Song K. Hyun SH, Shin T, Lee E. 2009. Post-activation treatment with demecolcine improves development of somatic cell nuclear transfer embryos in pigs by modifying the remodeling of donor nuclei. Mol. Reprod. Dev. 76: 611-619. https://doi.org/10.1002/mrd.20989
  41. Takahashi M, Keicho K, Takahashi H, Ogawa H, Schultz RM, Okano A. 2000. Effect of oxidative stress on development and DNA damage in in vitro cultured bovine embryos by comet assay. Theriogenology 54: 137-145. https://doi.org/10.1016/S0093-691X(00)00332-0
  42. Tao Y, Xie H, Hong H, Chen X, Jang J, Xia G. 2005. Effects of nitric oxide synthase inhibitors on porcine oocyte meiotic maturation. Zygote 13: 1-9. https://doi.org/10.1017/S0967199404002953
  43. Torner H, Brüssow KP, Alm H, Ratky J, Pöhland R, Tuchscherer A, Kanitz W. 2004. Mitochondrial aggregation n patterns and activity in porcine oocytes and apoptosis in surrounding cumulus cells depends on the stage of pre-ovulatory maturation, Theriogenology 61: 1675-1689. https://doi.org/10.1016/j.theriogenology.2003.09.013
  44. Van Blerkom J. 2008. Mitochondria as regulatory forces in oocytes, preimplantation embryos and stem cells. Reprod. Biomed. Online 16: 553-569. https://doi.org/10.1016/S1472-6483(10)60463-4
  45. Vargas F, Moreno JM, Wangensteen R, Rodriguez-Gomez I, Garcia-Estan J. 2007. The endocrine system in chronic nitric oxide deficiency. European Journal of Endocrinology 156: 1-12. https://doi.org/10.1530/eje.1.02314
  46. Walker SC, Shin T, Zaunbrecher GM, Romano JE, Johnson GA, Bazer FW, Piedrahita JA. 2002. A highly efficient method for porcine cloning by nuclear transfer using in vitro-matured oocytes. Cloning Stem cells 4: 105-112. https://doi.org/10.1089/153623002320253283
  47. Whitworth KM, Li R, Spate LD, Wax DM, Rieke A, Whyte JJ, Manandhar G, Sutovsky M, Green JA, Sutovsky P, Prather RS. 2009. Method of oocyte activation affects cloning efficiency in pigs. Mol. Reprod. Dev. 76: 490-500. https://doi.org/10.1002/mrd.20987
  48. Yoshioka K, Suzuki C, Tanaka A, Anas IM, Iwamura S. 2002. Birth of piglets derived from porcine zygotes cultured in a chemically defined medium. Biol. Reprod. 66: 112-119. https://doi.org/10.1095/biolreprod66.1.112
  49. You J, Lee J, Hyun SH, Lee E. 2012. L-carnitine treatment during oocyte maturation improves in vitro development of cloned pig embryos by influencing intracellular glutathione synthesis and embryonic gene expression. Theriogenology 78: 235-243. https://doi.org/10.1016/j.theriogenology.2012.02.027
  50. Zhang Y, Hogg N. 2005. S-Nitrosothiols: cellular formation and transport. Free Radic. Biol. Med. 38: 831-838. https://doi.org/10.1016/j.freeradbiomed.2004.12.016