Hostname: page-component-8448b6f56d-qsmjn Total loading time: 0 Render date: 2024-04-24T04:46:14.380Z Has data issue: false hasContentIssue false

Establishment of mouse androgenetic embryonic stem cells by double sperm injection and differentiation into beating embryoid body

Published online by Cambridge University Press:  23 September 2019

Lei Lei*
Affiliation:
Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, Heilongjiang, China Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, China
Lili Hu
Affiliation:
Department of Reproductive Medicine, the First People’s Hospital of Jining City, Jining, 272100, Shandong, China
Tong Li
Affiliation:
Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
Xinghui Shen
Affiliation:
Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
Xiao Liang
Affiliation:
Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang, China
Yajun Chen
Affiliation:
Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
Xiuqing Feng
Affiliation:
Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
Zhiwen Yang
Affiliation:
Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
Zhiyan Shan
Affiliation:
Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
*
Address for correspondence: Lei Lei. Department of Histology and Embryology, Harbin Medical University, 194 Xuefu Road, Nangang District, Harbin 150081, Heilongjiang, China, Tel: +86 451 86674518. Fax: +86 451 87503325. E-mail: lei086@ems.hrbmu.edu.cn

Summary

Androgenetic embryonic stem (AgES) cells offer a possible tool for patient-specific pluripotent stem cells that will benefit genomic imprinting studies and clinic applications. However, the difficulty in producing androgenetic embryos and the unbalanced expression of imprinted genes make the therapeutic applicability of AgES cells uncertain. In this study, we produced androgenetic embryos by injecting two sperm into an enucleated metaphase II (MII) oocyte. By this method, 88.48% of oocytes survived after injection, and 20.24% of these developed to the blastocyst stage. We successfully generated AgES cell lines from the androgenetic embryos and assayed the expression of imprinted genes in the cell lines. We found that the morphological characteristics of AgES cells were similar to that of fertilized embryonic stem cells (fES), such as expression of key pluripotent markers, and generation of cell derivatives representing all three germ layers following in vivo and in vitro differentiation. Furthermore, activation of paternal imprinted genes was detected, H19, ASC12 and Tss3 in AgES cell activation levels were lower while other examined genes showed no significant difference to that of fES cells. Interestingly, among examined maternal imprinted genes, only Mest and Igf2 were significantly increased, while levels of other detected genes were no different to that of fES cells. These results demonstrated that activation of some paternal imprinted genes, as well as recovery of maternal imprinted genes, was present in AgES cells. We differentiated AgES cells into a beating embryoid body in vitro, and discovered that the AgES cells did not show significant higher efficiency in myocardial differentiation potential.

Type
Research Article
Copyright
© Cambridge University Press 2019 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Allen, ND, Barton, SC, Hilton, K, Norris, ML and Surani, MA (1994) A functional analysis of imprinting in parthenogenetic embryonic stem cells. Development 120, 1473–82.Google ScholarPubMed
Barad, L, Schick, R, Zeevi-Levin, N, Itskovitz-Eldor, J and Binah, O (2014) Human embryonic stem cells vs human induced pluripotent stem cells for cardiac repair. Can J Cardiol 30, 1279–87.CrossRefGoogle ScholarPubMed
Beygo, J, Mertel, C, Kaya, S, Gillessen-Kaesbach, G, Eggermann, T, Horsthemke, B and Buiting, K (2018) The origin of imprinting defects in Temple syndrome and comparison with other imprinting disorders. Epigenetics 13, 822–8.CrossRefGoogle ScholarPubMed
Chen, FK, McLenachan, S, Edel, M, Da Cruz, L, Coffey, PJ and Mackey, DA (2014) iPS cells for modelling and treatment of retinal diseases. J Clin Med 3, 1511–41.CrossRefGoogle ScholarPubMed
Chen, W, Huang, Q, Ma, S and Li, M (2019) Progress in dopaminergic cell replacement and regenerative strategies for Parkinson’s disease. ACS Chem Neurosci 10, 839–51.CrossRefGoogle ScholarPubMed
Dinger, TC, Eckardt, S, Choi, SW, Camarero, G, Kurosaka, S, Hornich, V, McLaughlin, KJ and Muller, AM (2008) Androgenetic embryonic stem cells form neural progenitor cells in vivo and in vitro . Stem Cells 26, 1474–83.CrossRefGoogle ScholarPubMed
Eckardt, S, Leu, NA, Bradley, HL, Kato, H, Bunting, KD and McLaughlin, KJ (2007) Hematopoietic reconstitution with androgenetic and gynogenetic stem cells. Genes Dev 21, 409–19.CrossRefGoogle ScholarPubMed
Efstratiadis, A (1994) Parental imprinting of autosomal mammalian genes. Curr Opin Genet Dev 4, 265–80.CrossRefGoogle ScholarPubMed
Hu, JF, Vu, TH and Hoffman, AR (1997) Genomic deletion of an imprint maintenance element abolishes imprinting of both insulin-like growth factor II and H19. J Biol Chem 272, 20715–20.CrossRefGoogle ScholarPubMed
Hu, LL, Shen, XH, Zheng, Z, Wang, ZD, Liu, ZH, Jin, LH and Lei, L (2012) Cytochalasin B treatment of mouse oocytes during intracytoplasmic sperm injection (ICSI) increases embryo survival without impairment of development. Zygote 20, 361–9.CrossRefGoogle ScholarPubMed
Jelinic, P and Shaw, P (2007) Loss of imprinting and cancer. J Pathol 211, 261–8.CrossRefGoogle ScholarPubMed
Khosla, S, Dean, W, Brown, D, Reik, W and Feil, R (2001) Culture of preimplantation mouse embryos affects fetal development and the expression of imprinted genes. Biol Reprod 64, 918–26.CrossRefGoogle ScholarPubMed
Li, ZK, Wang, LY, Wang, LB, Feng, GH, Yuan, XW, Liu, C, Xu, K, Li, YH, Wan, HF, Zhang, Y, Li, YF, Li, X, Li, W, Zhou, Q and Hu, BY (2018) Generation of bimaternal and bipaternal mice from hypomethylated haploid ESCs with imprinting region deletions. Cell Stem Cell 23, 665–76, e664.CrossRefGoogle ScholarPubMed
Mann, JR and Stewart, CL (1991) Development to term of mouse androgenetic aggregation chimeras. Development 113, 1325–33.Google ScholarPubMed
McLaughlin, KJ, Kochanowski, H, Solter, D, Schwarzkopf, G, Szabo, PE and Mann, JR (1997) Roles of the imprinted gene Igf2 and paternal duplication of distal chromosome 7 in the perinatal abnormalities of androgenetic mouse chimeras. Development 124, 4897–904.Google ScholarPubMed
Menheniott, TR, Woodfine, K, Schulz, R, Wood, AJ, Monk, D, Giraud, AS, Baldwin, HS, Moore, GE and Oakey, RJ (2008) Genomic imprinting of Dopa decarboxylase in heart and reciprocal allelic expression with neighboring Grb10. Mol Cell Biol 28, 386–96.CrossRefGoogle ScholarPubMed
Miki, H, Hirose, M, Ogonuki, N, Inoue, K, Kezuka, F, Honda, A, Mekada, K, Hanaki, K, Iwafune, H, Yoshiki, A, Ishino, F and Ogura, A (2009) Efficient production of androgenetic embryos by round spermatid injection. Genesis 47, 155–60.CrossRefGoogle ScholarPubMed
Nativio, R, Sparago, A, Ito, Y, Weksberg, R, Riccio, A and Murrell, A (2011) Disruption of genomic neighbourhood at the imprinted IGF2-H19 locus in Beckwith–Wiedemann syndrome and Silver–Russell syndrome. Hum Mol Genet 20, 1363–74.CrossRefGoogle ScholarPubMed
Obata, Y, Ono, Y, Akuzawa, H, Kwon, OY, Yoshizawa, M and Kono, T (2000) Post-implantation development of mouse androgenetic embryos produced by in-vitro fertilization of enucleated oocytes. Hum Reprod 15, 874–80.CrossRefGoogle ScholarPubMed
Szabo, P and Mann, JR (1994) Expression and methylation of imprinted genes during in vitro differentiation of mouse parthenogenetic and androgenetic embryonic stem cell lines. Development 120, 1651–60.Google ScholarPubMed
Thomson, JA and Solter, D (1988) The developmental fate of androgenetic, parthenogenetic, and gynogenetic cells in chimeric gastrulating mouse embryos. Genes Dev 2, 1344–51.CrossRefGoogle ScholarPubMed
Zhao, Q, Wang, J, Zhang, Y, Kou, Z, Liu, S and Gao, S (2010) Generation of histocompatible androgenetic embryonic stem cells using spermatogenic cells. Stem Cells 28, 229–39.Google ScholarPubMed
Zhong, C, Yin, Q, Xie, Z, Bai, M, Dong, R, Tang, W, Xing, YH, Zhang, H, Yang, S, Chen, LL, Bartolomei, MS, Ferguson-Smith, A, Li, D, Yang, L, Wu, Y and Li, J (2015) CRISPR-Cas9-mediated genetic screening in mice with haploid embryonic stem cells carrying a guide rna library. Cell Stem Cell 17, 221–32.CrossRefGoogle ScholarPubMed
Supplementary material: File

Lei et al. supplementary material

Figure S1

Download Lei et al. supplementary material(File)
File 82 KB