DOI QR코드

DOI QR Code

Improved Preimplantation Development of Cloned Porcine Embryos through Supplementation of Histone Deacetylase Inhibitor MS-275

  • Fang, Xun (College of Veterinary Medicine, Chungnam National University) ;
  • Qamar, Ahmad Yar (College of Veterinary Medicine, Chungnam National University) ;
  • Shin, Sang Tae (College of Veterinary Medicine, Chungnam National University) ;
  • Cho, Jongki (College of Veterinary Medicine, Chungnam National University)
  • Received : 2019.09.18
  • Accepted : 2019.10.11
  • Published : 2019.10.31

Abstract

The objective of this study was to analyse the effects of MS-275 (Class I and II histone deacetylase inhibitor) supplementation on the development of porcine in-vitro somatic nuclear transfer embryo production. During in-vitro development, early embryos were exposed to different concentrations of MS-275 (0, $5{\mu}M$, $10{\mu}M$, and $20{\mu}M$). In in-vitro culture supplemented group, the blastocyst development rate was significantly enhanced by $10{\mu}M$ concentration than other groups (24.0% vs. 19.3%, 21.8%, 11.5%; P < 0.05). Additionally, the 6 h supplementation group, significantly improved the blastocysts production than 24 h, 48 h and control groups (26.1% vs. 17.0%, 15.2%, 2.8%; P < 0.05). Following supplementation with optimal concentrations and time ($10{\mu}M$-6 h group), the blastocyst production was significantly higher than control (25.7% vs 15.8%; P < 0.05). The optimal concentrations of MS-275 significantly enhanced the percentages of ICM:TE than control (43.6% vs. 38.4%; P < 0.05) accompanied with significantly higher expression levels of reprogramming related genes (POU5F1, Naong, and SOX2). In conclusion, the optimal concentrations of $10{\mu}M$ MS-275 and 6 h supplementation during in-vitro culture can significantly improve the quality of porcine in-vitro somatic nuclear transfer embryos through histone acetylation and epigenetic modification. Increasing the efficiency of clonal animal production will greatly promote the development of animal disease models and xenotransplantation.

Keywords

References

  1. Bavister BD, Leibfried ML, Lieberman G. Development of preimplantation embryos of the golden hamster in a defined culture medium. Biol Reprod 1983; 28: 235-247. https://doi.org/10.1095/biolreprod28.1.235
  2. Bo F, Di L, Qing-chang F, Liang R, Hong M, Liang W, Zhenhua G, Zhong-qiu L. Effect of trichostatin A on transfected donor cells and subsequent development of porcine cloned embryos. Zygote 2011; 19: 237-243. https://doi.org/10.1017/S0967199410000201
  3. Buemo CP, Gambini A, Moro LN, Hiriart MI, Fernandez-Martin R, Collas P, Salamone DF. Embryo aggregation in pig improves cloning efficiency and embryo quality. PLoS One 2016; 11: e0146390. https://doi.org/10.1371/journal.pone.0146390
  4. Cao Z, Li Y, Chen Z, Wang H, Zhang M, Zhou N, Wu R, Ling Y, Fang F, Li N, Zhang Y. Genome-wide dynamic profiling of histone methylation during nuclear transfer-mediated porcine somatic cell reprogramming. PLoS One 2015; 10: e0144897. https://doi.org/10.1371/journal.pone.0144897
  5. Cervera RP, Marti-Gutierrez N, Escorihuela E, Moreno R, Stojkovic M. Trichostatin A affects histone acetylation and gene expression in porcine somatic cell nucleus transfer embryos. Theriogenology 2009; 72: 1097-1110. https://doi.org/10.1016/j.theriogenology.2009.06.030
  6. Deshmukh RS, Ostrup O, Ostrup E, Vejlsted M, Niemann H, Lucas-Hahn A, Petersen B, Li J, Callesen H, Hyttel P. DNA methylation in porcine preimplantation embryos developed in vivo and produced by in vitro fertilization, parthenogenetic activation and somatic cell nuclear transfer. Epigenetics 2011; 6: 177-187. https://doi.org/10.4161/epi.6.2.13519
  7. du Puy L, Lopes SM, Haagsman HP, Roelen BA. Analysis of co-expression of OCT4, NANOG and SOX2 in pluripotent cells of the porcine embryo, in vivo and in vitro. Theriogenology 2011; 75: 513-526. https://doi.org/10.1016/j.theriogenology.2010.09.019
  8. Fang X, Qamar AY, Yoon KY, Cho JK. Improved preimplantation development of porcine cloned embryos by flavone supplement as antioxidant. J Emb Trans 2018; 33: 255-264. https://doi.org/10.12750/JET.2018.33.4.255
  9. Hassan BMS, Fang X, Roy PK, Shin ST, Cho JK. Effect of alpha lipoic acid as an antioxidant supplement during in vitro maturation medium on bovine embryonic development. J Emb Trans 2017; 32: 123-130. https://doi.org/10.12750/JET.2017.32.3.123
  10. Holm IE, Alstrup AK, Luo Y. Genetically modified pig models for neurodegenerative disorders. J Pathol 2016; 238: 267-287. https://doi.org/10.1002/path.4654
  11. Huang Y, Yuan L, Li T, Wang A, Li Z, Pang D, Wang B, Ouyang H. Valproic acid improves porcine parthenogenetic embryo development through transient remodeling of histone modifiers. Cell Physiol Biochem 2015; 37: 1463-1473. https://doi.org/10.1159/000438515
  12. Hwang JH, Kim SE, Gupta MK, Lee H. Gnotobiotic miniature pig interbreed somatic cell nuclear transfer for xenotransplantation. Cell Reprogram 2016; 18: 207-213. https://doi.org/10.1089/cell.2015.0065
  13. Jin JX, Lee S, Taweechaipaisankul A, Kim GA, Lee BC. The HDAC inhibitor LAQ824 enhances epigenetic reprogramming and in vitro development of porcine SCNT embryos. Cell Physiol Biochem 2017; 41: 1255-1266. https://doi.org/10.1159/000464389
  14. Jin L, Guo Q, Zhang GL, Xing XX, Xuan MF, Luo QR, Luo ZB, Wang JX, Yin XJ, Kang JD. The histone deacetylase inhibitor, CI994, improves nuclear reprogramming and in vitro developmental potential of cloned pig embryos. Cell Reprogram 2018; 20: 205-213. https://doi.org/10.1089/cell.2018.0001
  15. Keefer CL. Artificial cloning of domestic animals. Proc Natl Acad Sci U S A 2015; 112: 8874-8878. https://doi.org/10.1073/pnas.1501718112
  16. Kim G, Roy PK, Hassan BMS, Cho JK. Improved preimplantation development of porcine somatic cell nuclear transfer embryos by caffeine treatment. J Vet Sci 2019; 20: e31. https://doi.org/10.4142/jvs.2019.20.e31
  17. Lagutina I, Fulka H, Lazzari G, Galli C. Interspecies somatic cell nuclear transfer: advancements and problems. Cell Reprogram 2013; 15: 374-384. https://doi.org/10.1089/cell.2013.0036
  18. Ma S, Liu T, Xu L, Wang Y, Zhou J, Huang T, Li P, Liu H, Zhang Y, Zhou X, Cui Y, Zang X, Wang Y, Guan F. Histone deacetylases inhibitor MS-275 suppresses human esophageal squamous cell carcinoma cell growth and progression via the PI3K/Akt/mTOR pathway. J Cell Physiol 2019; 12: 22400-22410.
  19. Malaweera DBO, Ramachandra S, Wu JB, Oh SK, Kim SH, Kim SJ, Shin ST, Cho JK. Establishment of efficient microinjection system in the porcine embryos. J Emb Trans 2014; 29: 59-66. https://doi.org/10.12750/JET.2014.29.1.59
  20. Mao J, Zhao MT, Whitworth KM, Spate LD, Walters EM, O'Gorman C, Lee K, Samuel MS, Murphy CN, Wells K, Rivera RM, Prather RS. Oxamflatin treatment enhances cloned porcine embryo development and nuclear reprogramming. Cell Reprogram 2015; 17: 28-40. https://doi.org/10.1089/cell.2014.0075
  21. Matoba S, Zhang Y. Somatic Cell Nuclear Transfer Reprogramming: Mechanisms and Applications. Cell Stem Cell 2018; 23: 471-485. https://doi.org/10.1016/j.stem.2018.06.018
  22. Min KH, Na SW, Lee EC, Kim GY, Fang X, Hassan BMS, Cho JK. Study on Chemicals for post-activation in porcine somatic cell nuclear transfer. J Emb Trans 2016; 31: 131-136. https://doi.org/10.12750/JET.2016.31.2.131
  23. Na SW, Lee EC, Kim GY, Min KH, Yu YK, Roy PK, Fang X, Hassan BMS, Yoon KY, Shin ST, Cho JK. Study on embryo transfer system for production of transgenic pigs. J Emb Trans 2015; 30: 345-350. https://doi.org/10.12750/JET.2015.30.4.345
  24. Nalawansha DA, Gomes ID, Wambua MK, Pflum MKH. HDAC inhibitor-induced mitotic arrest is mediated by Eg5/KIF11 acetylation. Cell Chem Biol 2017; 24: 481-492. https://doi.org/10.1016/j.chembiol.2017.03.008
  25. Oh SK, Malaweera DBO, Ramachandra S, Shin ST, Cho JK. Effect of 7,8-dihydroxyflavone on in vitro maturation of oocytes in pigs. J Emb Trans 2014; 29: 67-72. https://doi.org/10.12750/JET.2014.29.1.67
  26. Pasque V, Jullien J, Miyamoto K, Halley-Stott RP, Gurdon JB. Epigenetic factors influencing resistance to nuclear reprogramming. Trends Genet 2011; 27: 516-525. https://doi.org/10.1016/j.tig.2011.08.002
  27. Roy PK, Fang X, Hassan BMS, Shin ST, Cho JK. Effects of roscovitine on in vitro development of porcine oocyte using brilliant cresyl blue. J Emb Trans 2017; 32: 111-122. https://doi.org/10.12750/JET.2017.32.3.111
  28. Roy PK, Kim KY, Fang X, Hassan BMS, De Zoysa M, Shin ST, Cho JK. Optimization of post-activation systems to improve the embryonic development in porcine parthenogenesis and somatic cell nuclear transfer. J Emb Trans 2017; 32: 95-104. https://doi.org/10.12750/JET.2017.32.3.95
  29. Rybouchkin A, Kato Y, Tsunoda Y. Role of histone acetylation in reprogramming of somatic nuclei following nuclear transfer. Biol Reprod 2006; 74: 1083-1089. https://doi.org/10.1095/biolreprod.105.047456
  30. Simmet K, Zakhartchenko V, Philippou-Massier J, Blum H, Klymiuk N, Wolf E. OCT4/POU5F1 is required for NANOG expression in bovine blastocysts. Proc Natl Acad Sci U S A 2018; 115: 2770-2775. https://doi.org/10.1073/pnas.1718833115
  31. Song Y, Hai T, Wang Y, Guo R, Li W, Wang L, Zhou Q. Epigenetic reprogramming, gene expression and in vitro development of porcine SCNT embryos are significantly improved by a histone deacetylase inhibitor--m-carboxycinnamic acid bishydroxamide (CBHA). Protein Cell 2014; 5: 382-293. https://doi.org/10.1007/s13238-014-0034-3
  32. Taweechaipaisankul A, Jin JX, Lee S, Kim GA, Suh YH, Ahn MS, Park SJ, Lee BY, Lee BC. Improved early development of porcine cloned embryos by treatment with quisinostat, a potent histone deacetylase inhibitor. J Reprod Dev 2019; 65: 103-112. https://doi.org/10.1262/jrd.2018-098
  33. Wang H, Cui W, Meng C, Zhang J, Li Y, Qian Y, Xing G, Zhao D, Cao S. MC1568 enhances histone acetylation during oocyte meiosis and improves development of somatic cell nuclear transfer embryos in pig. Cell Reprogram 2018; 20: 55-65. https://doi.org/10.1089/cell.2017.0023
  34. Whitworth KM, Mao J, Lee K, Spollen WG, Samuel MS, Walters EM, Spate LD, Prather RS. Transcriptome analysis of pig in vivo, in vitro-fertilized, and nuclear transfer blastocyststage embryos treated with histone deacetylase inhibitors postfusion and activation reveals changes in the lysosomal pathway. Cell Reprogram 2015; 17: 243-258. https://doi.org/10.1089/cell.2015.0022
  35. Yin Y, Hao H, Xu X, Shen L, Wu W, Zhang J, Li Q. Generation of an MC3R knock-out pig by CRSPR/Cas9 combined with somatic cell nuclear transfer (SCNT) technology. Lipids Health Dis 2019; 18: 122. https://doi.org/10.1186/s12944-019-1073-9
  36. You J, Lee J, Hyun SH, Lee E. L-carnitine treatment during oocyte maturation improves in vitro development of cloned pig embryos by influencing intracellular glutathione synthesis and embryonic gene expression. Theriogenology 2012; 78: 235-243. https://doi.org/10.1016/j.theriogenology.2012.02.027
  37. Yu YK, Roy PK, Min KH, Na SW, Lee EC, Kim GY, Fang X, Hassan BMS, Cho JK. Effect of insulin supplement on development of porcine parthenogenetic embryos. J Emb Trans 2016; 31: 123-129. https://doi.org/10.12750/JET.2016.31.2.123
  38. Zhao J, Whyte J, Prather RS. Effect of epigenetic regulation during swine embryogenesis and on cloning by nuclear transfer. Cell Tissue Res 2010; 341: 13-21. https://doi.org/10.1007/s00441-010-1000-x

Cited by

  1. Chitosan nanoparticles enhance developmental competence of in vitro‐matured porcine oocytes vol.56, pp.2, 2019, https://doi.org/10.1111/rda.13871