Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

Effects of dimethyl sulfoxide (DMSO) on DNA methylation and histone modification in parthenogenetically activated porcine embryos

Hui Cheng A # , Yu Han A # , Jian Zhang A , Sheng Zhang B , Yanhui Zhai A , Xinglan An B , Qi Li B , Jiahui Duan A , Xueming Zhang A , Ziyi Li B , Bo Tang https://orcid.org/0000-0002-6033-8199 A * and Haiqing Shen A *
+ Author Affiliations
- Author Affiliations

A College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China.

B Academy of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin 130021, China.

# These authors contributed equally to this paper

Handling Editor: Ye Yuan

Reproduction, Fertility and Development 34(8) 598-607 https://doi.org/10.1071/RD21083
Published online: 11 April 2022

© 2022 The Author(s) (or their employer(s)). Published by CSIRO Publishing

Abstract

Epigenetic mechanisms play an important role in oogenesis and early embryo development in mammals. Dimethyl sulfoxide (DMSO) is frequently used as a solvent in biological studies and as a vehicle for drug therapy. Recent studies suggest that DMSO detrimentally affects porcine embryonic development, yet the mechanism of the process in parthenogenetically activated porcine embryos has not been reported. In this study, we found that treatment of embryos with 1.5% DMSO significantly decreased the cleavage and blastocyst rates, total cell number of blastocysts and the anti-apoptotic gene BCL-2 transcription level; however, the percentage of apoptotic cells and the expression levels of the pro-apoptotic gene BAX were not changed. Treatment with DMSO significantly decreased the expression levels of DNMT1, DNMT3a, DNMT3b, TET1, TET2, TET3, KMT2C, MLL2 and SETD3 in most of the stages of embryonic development and increased 5-mC signals, while the staining intensity for 5-hmC had no change in porcine preimplantation embryos from 2-cell to the blastocyst stages. Meanwhile, DMSO decreased the level of H3K4me3 during the development of parthenogenetically activated porcine embryos. After treatment with DMSO, expression levels of the pluripotency-related genes POU5F1 and NANOG decreased significantly (P < 0.01), whereas the imprinted gene H19 did not change (P > 0.05). In conclusion, these results suggest that DMSO can affect genome-wide DNA methylation and histone modification by regulating the expression of epigenetic modification enzymes, and DMSO also influences the expression level of pluripotent genes. These dysregulations lead to defects in embryonic development.

Keywords: DMSO, DNA methylation, embryonic development, gene, histone modification, parthenogenetically activated, porcine, preimplantation embryos.


References

Arai, Y, Umeyama, K, Takeuchi, K, Okazaki, N, Hichiwa, N, Yashima, S, Nakano, K, Nagashima, H, and Ohgane, J (2017). Establishment of DNA methylation patterns of the Fibrillin1 (FBN1) gene in porcine embryos and tissues. Journal of Reproduction and Development 63, 157–165.
Establishment of DNA methylation patterns of the Fibrillin1 (FBN1) gene in porcine embryos and tissues.Crossref | GoogleScholarGoogle Scholar |

Barlow, DP, and Bartolomei, MS (2014). Genomic imprinting in mammals. Cold Spring Harbor Perspectives in Biology 6, a018382.
Genomic imprinting in mammals.Crossref | GoogleScholarGoogle Scholar | 24492710PubMed |

Bartolomei, MS, and Ferguson-Smith, AC (2011). Mammalian genomic imprinting. Cold Spring Harbor Perspectives in Biology 3, a002592.
Mammalian genomic imprinting.Crossref | GoogleScholarGoogle Scholar | 21576252PubMed |

Brevini, TAL, and Gandolfi, F (2008). Parthenotes as a source of embryonic stem cells. Cell Proliferation 41, 20–30.

Cheng, H, Wang, Y, Zhang, J, Zhang, S, Ma, X, An, X, Man, X, Zhang, X, Li, Z, and Tang, B (2018). Effects of PRDM14 silencing on parthenogenetically activated porcine embryos. Cellular Reprogramming 20, 382–388.
Effects of PRDM14 silencing on parthenogenetically activated porcine embryos.Crossref | GoogleScholarGoogle Scholar | 30325654PubMed |

Cheng, H, Zhang, J, Zhang, S, Zhai, Y, Jiang, Y, An, X, Ma, X, Zhang, X, Li, Z, and Tang, B (2019). Tet3 is required for normal in vitro fertilization preimplantation embryos development of bovine. Molecular Reproduction and Development 86, 298–307.
Tet3 is required for normal in vitro fertilization preimplantation embryos development of bovine.Crossref | GoogleScholarGoogle Scholar | 30624819PubMed |

Chetty, S, Pagliuca, FW, Honore, C, Kweudjeu, A, Rezania, A, and Melton, DA (2013). A simple tool to improve pluripotent stem cell differentiation. Nature Methods 10, 553–556.
A simple tool to improve pluripotent stem cell differentiation.Crossref | GoogleScholarGoogle Scholar | 23584186PubMed |

Choi, T (2011). Dimethyl sulfoxide inhibits spontaneous oocyte fragmentation and delays inactivation of maturation promoting factor (MPF) during the prolonged culture of ovulated murine oocytes in vitro. Cytotechnology 63, 279–284.
Dimethyl sulfoxide inhibits spontaneous oocyte fragmentation and delays inactivation of maturation promoting factor (MPF) during the prolonged culture of ovulated murine oocytes in vitro.Crossref | GoogleScholarGoogle Scholar | 21336963PubMed |

Choi, S-C, Choi, J-H, Cui, L-H, Seo, H-R, Kim, J-H, Park, C-Y, Joo, H-J, Park, J-H, Hong, S-J, Yu, C-W, and Lim, D-S (2015). Mixl1 and Flk1 are key players of Wnt/TGF-β signaling during DMSO-induced mesodermal specification in P19 cells. Journal of Cellular Physiology 230, 1807–1821.
Mixl1 and Flk1 are key players of Wnt/TGF-β signaling during DMSO-induced mesodermal specification in P19 cells.Crossref | GoogleScholarGoogle Scholar | 25521758PubMed |

Edwards, JR, Yarychkivska, O, Boulard, M, and Bestor, TH (2017). DNA methylation and DNA methyltransferases. Epigenetics & Chromatin 10, 23.
DNA methylation and DNA methyltransferases.Crossref | GoogleScholarGoogle Scholar |

Godini, R, Lafta, HY, and Fallahi, H (2018). Epigenetic modifications in the embryonic and induced pluripotent stem cells. Gene Expression Patterns 29, 1–9.
Epigenetic modifications in the embryonic and induced pluripotent stem cells.Crossref | GoogleScholarGoogle Scholar | 29625185PubMed |

Gook, DA, Choo, B, Bourne, H, Lewis, K, and Edgar, DH (2016). Closed vitrification of human oocytes and blastocysts: outcomes from a series of clinical cases. Journal of Assisted Reproduction and Genetics 33, 1247–1252.
Closed vitrification of human oocytes and blastocysts: outcomes from a series of clinical cases.Crossref | GoogleScholarGoogle Scholar | 27233650PubMed |

Hou, X, Liu, J, Zhang, Z, Zhai, Y, Wang, Y, Wang, Z, Tang, B, Zhang, X, Sun, L, and Li, Z (2016). Effects of cytochalasin B on DNA methylation and histone modification in parthenogenetically activated porcine embryos. Reproduction 152, 519–527.
Effects of cytochalasin B on DNA methylation and histone modification in parthenogenetically activated porcine embryos.Crossref | GoogleScholarGoogle Scholar | 27581081PubMed |

Huang, J, Zhang, H, Wang, X, Dobbs, KB, Yao, J, Qin, G, Whitworth, K, Walters, EM, Prather, RS, and Zhao, J (2015). Impairment of preimplantation porcine embryo development by histone demethylase KDM5B knockdown through disturbance of bivalent H3K4me3-H3K27me3 modifications. Biology of Reproduction 92, 72, 1–11.
Impairment of preimplantation porcine embryo development by histone demethylase KDM5B knockdown through disturbance of bivalent H3K4me3-H3K27me3 modifications.Crossref | GoogleScholarGoogle Scholar |

Kang, M-H, Das, J, Gurunathan, S, Park, H-W, Song, H, Park, C, and Kim, J-H (2017). The cytotoxic effects of dimethyl sulfoxide in mouse preimplantation embryos: a mechanistic study. Theranostics 7, 4735–4752.
The cytotoxic effects of dimethyl sulfoxide in mouse preimplantation embryos: a mechanistic study.Crossref | GoogleScholarGoogle Scholar | 29187900PubMed |

Kim, M, and Costello, J (2017). DNA methylation: an epigenetic mark of cellular memory. Experimental & Molecular Medicine 49, e322.
DNA methylation: an epigenetic mark of cellular memory.Crossref | GoogleScholarGoogle Scholar |

Kooistra, SM, and Helin, K (2012). Molecular mechanisms and potential functions of histone demethylases. Nature Reviews Molecular Cell Biology 13, 297–311.
Molecular mechanisms and potential functions of histone demethylases.Crossref | GoogleScholarGoogle Scholar | 22473470PubMed |

Li, X, Wang, Y-K, Song, Z-Q, Du, Z-Q, and Yang, C-X (2016). Dimethyl sulfoxide perturbs cell cycle progression and spindle organization in porcine meiotic oocytes. PLoS ONE 11, e0158074.
Dimethyl sulfoxide perturbs cell cycle progression and spindle organization in porcine meiotic oocytes.Crossref | GoogleScholarGoogle Scholar | 27348312PubMed |

Liao, J, Luo, S, Yang, M, and Lu, Q (2020). Overexpression of CXCR5 in CD4+ T cells of SLE patients caused by excessive SETD3. Clinical Immunology 214, 108406.
Overexpression of CXCR5 in CD4+ T cells of SLE patients caused by excessive SETD3.Crossref | GoogleScholarGoogle Scholar | 32240818PubMed |

Liu, L, Song, G, Gao, F, Guan, J, Tang, B, and Li, Z (2012). Transient exposure to sodium butyrate after germinal vesicle breakdown improves meiosis but not developmental competence in pig oocytes. Cell Biology International 36, 483–490.
Transient exposure to sodium butyrate after germinal vesicle breakdown improves meiosis but not developmental competence in pig oocytes.Crossref | GoogleScholarGoogle Scholar | 22288569PubMed |

Ma, S-F, Liu, X-Y, Miao, D-Q, Han, Z-B, Zhang, X, Miao, Y-L, Yanagimachi, R, and Tan, J-H (2005). Parthenogenetic activation of mouse oocytes by strontium chloride: a search for the best conditions. Theriogenology 64, 1142–1157.
Parthenogenetic activation of mouse oocytes by strontium chloride: a search for the best conditions.Crossref | GoogleScholarGoogle Scholar | 16125558PubMed |

Nohalez, A, Martinez, CA, Gil, MA, Almiñana, C, Roca, J, Martinez, EA, and Cuello, C (2015). Effects of two combinations of cryoprotectants on the in vitro developmental capacity of vitrified immature porcine oocytes. Theriogenology 84, 545–552.
Effects of two combinations of cryoprotectants on the in vitro developmental capacity of vitrified immature porcine oocytes.Crossref | GoogleScholarGoogle Scholar | 25998270PubMed |

Pal, R, Mamidi, MK, Das, AK, and Bhonde, R (2012). Diverse effects of dimethyl sulfoxide (DMSO) on the differentiation potential of human embryonic stem cells. Archives of Toxicology 86, 651–661.
Diverse effects of dimethyl sulfoxide (DMSO) on the differentiation potential of human embryonic stem cells.Crossref | GoogleScholarGoogle Scholar | 22105179PubMed |

Reik, W, Dean, W, and Walter, J (2001). Epigenetic reprogramming in mammalian development. Science 293, 1089–1093.
Epigenetic reprogramming in mammalian development.Crossref | GoogleScholarGoogle Scholar | 11498579PubMed |

Santos, NC, Figueira-Coelho, J, Martins-Silva, J, and Saldanha, C (2003). Multidisciplinary utilization of dimethyl sulfoxide: pharmacological, cellular, and molecular aspects. Biochemical Pharmacology 65, 1035–1041.
Multidisciplinary utilization of dimethyl sulfoxide: pharmacological, cellular, and molecular aspects.Crossref | GoogleScholarGoogle Scholar | 12663039PubMed |

Schulz, KN, and Harrison, MM (2019). Mechanisms regulating zygotic genome activation. Nature Reviews Genetics 20, 221–234.
Mechanisms regulating zygotic genome activation.Crossref | GoogleScholarGoogle Scholar | 30573849PubMed |

Shao, G-B, Chen, J-C, Zhang, L-P, Huang, P, Lu, H-Y, Jin, J, Gong, A-H, and Sang, J-R (2014). Dynamic patterns of histone H3 lysine 4 methyltransferases and demethylases during mouse preimplantation development. In Vitro Cellular & Developmental Biology - Animal 50, 603–613.
Dynamic patterns of histone H3 lysine 4 methyltransferases and demethylases during mouse preimplantation development.Crossref | GoogleScholarGoogle Scholar |

Shen, L, Inoue, A, He, J, Liu, Y, Lu, F, and Zhang, Y (2014). Tet3 and DNA replication mediate demethylation of both the maternal and paternal genomes in mouse zygotes. Cell Stem Cell 15, 459–471.
Tet3 and DNA replication mediate demethylation of both the maternal and paternal genomes in mouse zygotes.Crossref | GoogleScholarGoogle Scholar | 25280220PubMed |

Su, J, Wang, Y, Li, Y, Li, R, Li, Q, Wu, Y, Quan, F, Liu, J, Guo, Z, and Zhang, Y (2011). Oxamflatin significantly improves nuclear reprogramming, blastocyst quality, and in vitro development of bovine SCNT embryos. PLoS ONE 6, e23805.
Oxamflatin significantly improves nuclear reprogramming, blastocyst quality, and in vitro development of bovine SCNT embryos.Crossref | GoogleScholarGoogle Scholar | 21912607PubMed |

Terranova, R, Agherbi, H, Boned, A, Meresse, S, and Djabali, M (2006). Histone and DNA methylation defects at Hox genes in mice expressing a SET domain-truncated form of Mll. Proceedings of the National Academy of Sciences of the United States of America 103, 6629–6634.
Histone and DNA methylation defects at Hox genes in mice expressing a SET domain-truncated form of Mll.Crossref | GoogleScholarGoogle Scholar | 16618927PubMed |

Trowbridge, JJ, Snow, JW, Kim, J, and Orkin, SH (2009). DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells. Cell Stem Cell 5, 442–449.
DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells.Crossref | GoogleScholarGoogle Scholar | 19796624PubMed |

Uysal, F, Akkoyunlu, G, and Ozturk, S (2015). Dynamic expression of DNA methyltransferases (DNMTs) in oocytes and early embryos. Biochimie 116, 103–113.
Dynamic expression of DNA methyltransferases (DNMTs) in oocytes and early embryos.Crossref | GoogleScholarGoogle Scholar | 26143007PubMed |

Williams, K, Christensen, J, Pedersen, MT, Johansen, JV, Cloos, PAC, Rappsilber, J, and Helin, K (2011). TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature 473, 343–348.
TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity.Crossref | GoogleScholarGoogle Scholar | 21490601PubMed |

Zhai, Y, Li, W, Zhang, Z, Cao, Y, Wang, Z, Zhang, S, and Li, Z (2018). Epigenetic states of donor cells significantly affect the development of somatic cell nuclear transfer (SCNT) embryos in pigs. Molecular Reproduction and Development 85, 26–37.
Epigenetic states of donor cells significantly affect the development of somatic cell nuclear transfer (SCNT) embryos in pigs.Crossref | GoogleScholarGoogle Scholar | 29205617PubMed |

Zhou, D, Shen, X, Gu, Y, Zhang, N, Li, T, Wu, X, and Lei, L (2014). Effects of dimethyl sulfoxide on asymmetric division and cytokinesis in mouse oocytes. BMC Developmental Biology 14, 28.
Effects of dimethyl sulfoxide on asymmetric division and cytokinesis in mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 24953160PubMed |

Zhu, Q, Stöger, R, and Alberio, R (2018). A lexicon of DNA modifications: their roles in embryo development and the germline. Frontiers in Cell and Developmental Biology 6, 24.
A lexicon of DNA modifications: their roles in embryo development and the germline.Crossref | GoogleScholarGoogle Scholar | 29637072PubMed |