Genome-wide assessment of DNA methylation in mouse oocytes reveals effects associated with in vitro growth, superovulation, and sexual maturity

Maria Desemparats Saenz-de-Juano, Elena Ivanova, Katy Billooye, Anamaria-Cristina Herta, Johan Smitz, Gavin Kelsey, Ellen Anckaert, Maria Desemparats Saenz-de-Juano, Elena Ivanova, Katy Billooye, Anamaria-Cristina Herta, Johan Smitz, Gavin Kelsey, Ellen Anckaert

Abstract

Background: In vitro follicle culture (IFC), as applied in the mouse system, allows the growth and maturation of a large number of immature preantral follicles to become mature and competent oocytes. In the human oncofertility clinic, there is increasing interest in developing this technique as an alternative to ovarian cortical tissue transplantation and to preserve the fertility of prepubertal cancer patients. However, the effect of IFC and hormonal stimulation on DNA methylation in the oocyte is not fully known, and there is legitimate concern over epigenetic abnormalities that could be induced by procedures applied during assisted reproductive technology (ART).

Results: In this study, we present the first genome-wide analysis of DNA methylation in MII oocytes obtained after natural ovulation, after IFC and after superovulation. We also performed a comparison between prepubertal and adult hormonally stimulated oocytes. Globally, the distinctive methylation landscape of oocytes, comprising alternating hyper- and hypomethylated domains, is preserved irrespective of the procedure. The conservation of methylation extends to the germline differential methylated regions (DMRs) of imprinted genes, necessary for their monoallelic expression in the embryo. However, we do detect specific, consistent, and coherent differences in DNA methylation in IFC oocytes, and between oocytes obtained after superovulation from prepubertal compared with sexually mature females. Several methylation differences span entire transcription units. Among these, we found alterations in Tcf4, Sox5, Zfp521, and other genes related to nervous system development.

Conclusions: Our observations show that IFC is associated with altered methylation at specific set of loci. DNA methylation of superovulated prepubertal oocytes differs from that of superovulated adult oocytes, whereas oocytes from superovulated adult females differ very little from naturally ovulated oocytes. Importantly, we show that regions other than imprinted gDMRs are susceptible to methylation changes associated with superovulation, IFC, and/or sexual immaturity in mouse oocytes. Our results provide an important reference for the use of in vitro growth and maturation of oocytes, particularly from prepubertal females, in assisted reproductive treatments or fertility preservation.

Keywords: Global DNA methylation; In vitro follicle culture; Prepubertal oocytes; Superovulation.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
a Experimental groups used for the genome-wide DNA methylation analysis. The number of females used per group is indicated in brackets next to their age. The tubes represent the number of biological replicates and the number under each tube indicates the number of oocytes pooled in each sample. MII metaphase II, eCG equine chorionic gonadotropin, hCG human chorionic gonadotropin. b MII oocytes from IFC obtained after 10 days of culture. Pictures were taken before snap freezing to measure oocyte diameters. c Oocyte diameter per group. Bar charts show the mean and the standard deviation (SD). Lower-case letters denote significant differences (p < 0.05) after applying non-parametric Krustall-Wallis and Dunn’s multiple comparisons tests
Fig. 2
Fig. 2
a Distribution of DNA methylation across the genome in 100-CpG windows in all samples compared to Germinal Vesicle (GV) oocytes from Shirane et al. [38]. b SeqMonk screenshot of a 1.7 Mb region of chromosome 11 depicting the hypermethylated (HyperD) and hypomethylated (HypoD) domains characteristic of the oocyte methylome in each of the 16 individual methylation datasets. Genes and oocyte mRNA are shown in red or blue depending on their direction of transcription (forward and reverse, respectively). Each color-coded vertical bar in the screenshot represents the methylation value of a non-overlapping 100 CpG tile. HypoD, HyperD, and oocyte mRNA annotation tracks are derived from Veselovska et al. [24]. c DNA methylation percentages at HyperD in all samples (n = 26,570). In the box:whiskers plot, the line across the middle of the box shows the median, the upper and lower extremities of the box show the 25th and 75th percentile of the set of data, and the upper and lower black whiskers show the median plus/minus the interquartile (25–75%) range multiplied by 2. Individual points which fall outside this range are shown as filled circles, and represent single outlier tiles. d Box:whisker plot showing the DNA methylation percentages at HypoD (n = 38,739). e DNA methylation percentages of CpG Islands (CGI) located at promoters (n = 11,542) and CGIs highly methylated in oocytes (n = 2014). Each point represents the mean value along with error bars indicating the 95% confidence interval for the measure. f Principal component analysis (PCA) of informative 100-CpG tiles (value between 0 and 100 in all 16 samples; n = 195,170) shows how biological replicates cluster together within each group and differently between conditions
Fig. 3
Fig. 3
a Scatterplot for informative tiles (100 CpG window size, n = 195,170) in both IFC and SO. Data from replicates are pooled. Differentially methylated tiles (p < 0.05) identified by logistic regression and with a methylation difference of ≥ 20% are highlighted in blue or red (hypomethylated in IFC and hypermethylated in IFC, respectively). b Heat map after unsupervised hierarchical clustering of all differentially methylated tiles (p < 0.05, 100-CpG window size, n = 6362) between IFC and SO. The heatmap shows how biological replicates were consistent within groups and IFC differed in a similar way from SO, SOA, and IV for these differentially methylated sites. c SeqMonk screenshot of a 6 Mbp region of chromosome 6 showing methylation at the Sox5 locus, with 18 hypomethylated tiles in IFC. Each color-coded vertical bar in the screenshot represents the methylation value of a non-overlapping 100-CpG tile. Genes and oocyte mRNA are shown in red or blue depending on their direction of transcription (forward or reverse, respectively)
Fig. 4
Fig. 4
a Scatterplot for common informative tiles (100 CpG window size, n = 195,170 between SO and SOA. Data from replicates are pooled. Differentially methylated tiles (p < 0.05) identified by logistic regression and with a methylation difference of ≥ 20% are highlighted in blue or red (hypomethylated in IFC and hypermethylated in IFC, respectively). b Heat map after unsupervised hierarchical clustering of all differentially methylated tiles (p < 0.05, 100 CpG window size, n = 14,795 between SO and SOA. The heatmap shows that the IFC group followed the same trend as SO, while the IV group was similar to SOA for these differentially methylated sites. c SeqMonk screenshot showing the methylation levels at Tcf4 locus (with 28 hypermethylated tiles). Each color-coded vertical bar represents the methylation value of a non-overlapping 100-CpG tile. Genes and oocyte mRNA are shown in red or blue depending on their direction of transcription (forward or reverse, respectively). d, e Venn diagrams showing the common hypomethylated and hypermethylated genes that were affected in IFC vs. SO, SO vs. SOA, and IFC vs. SOA
Fig. 5
Fig. 5
a SeqMonk screenshot of the DNA methylation profiles of the CGIs at the locus Sfi1 in chromosome 11. Each color-coded vertical bar represents the methylation value of a differentially methylated CGI. Genes and oocyte mRNA are shown in red or blue depending on their direction of transcription (forward or reverse, respectively). b Box-whisker plot showing the DNA methylation levels at 28 maternally imprinted germline differentially methylated regions (gDMRs) in each replicate. In the plots, the line across the middle of the box shows the median, the upper and lower extremities of the box show the 25th and 75th percentile of the set of data, and the upper and lower black whiskers show the median plus/minus the interquartile (25–75%) range multiplied by 2. Individual points that fall outside this range are shown as filled circles and represent single outlier tiles. c DNA methylation levels at the Nespas-Gnasxl gDMR for each sample. d SeqMonk screenshot of the DNA methylation distribution (100-CpG tiles quantified) in relation to the gene structure of Dnmt1. The data for the replicates is combined into the tracks labeled IFC, SO, SOA, and IV. Each color-coded bar represents the methylation value of a non-overlapping 100-CpG tile. The direction of transcription is represented by the arrows. The promoter of the oocyte transcript is marked with a black bar

References

    1. Hikabe O, Hamazaki N, Nagamatsu G, Obata Y, Hirao Y, Hamada N, et al. Reconstitution in vitro of the entire cycle of the mouse female germ line. Nature. 2016;539:299–303. doi: 10.1038/nature20104.
    1. Smitz JEJ, Gilchrist RB. Are human oocytes from stem cells next? Nat Biotechnol. 2016;34:1247–1248. doi: 10.1038/nbt.3742.
    1. Sánchez F, Lolicato F, Romero S, De Vos M, Van Ranst H, Verheyen G, et al. An improved IVM method for cumulus-oocyte complexes from small follicles in polycystic ovary syndrome patients enhances oocyte competence and embryo yield. Hum Reprod. 2017;32:2056–2068. doi: 10.1093/humrep/dex262.
    1. Laronda MM, Rutz AL, Xiao S, Whelan KA, Duncan FE, Roth EW, et al. A bioprosthetic ovary created using 3D printed microporous scaffolds restores ovarian function in sterilized mice. Nat Commun. 2017;8:1–10. doi: 10.1038/ncomms15261.
    1. Xiao S, Coppeta JR, Rogers HB, Isenberg BC, Zhu J, Olalekan SA, et al. A microfluidic culture model of the human reproductive tract and 28-day menstrual cycle. Nat Commun. 2017;8:1–13. doi: 10.1038/s41467-016-0009-6.
    1. Xiao S, Zhang J, Romero MM, Smith KN, Shea LD, Woodruff TK. In vitro follicle growth supports human oocyte meiotic maturation. Sci Rep. 2015;5:1–5.
    1. McLaughlin M, Albertini DF, Wallace WHB, Anderson RA, Telfer EE. Metaphase II oocytes from human unilaminar follicles grown in a multi-step culture system. MHR Basic Sci Reprod Med. 2018;24(3):1–8.
    1. Canovas S, Ross PJ, Kelsey G, Coy P. DNA methylation in embryo development: epigenetic impact of ART (assisted reproductive technologies) BioEssays. 2017;39:1–11. doi: 10.1002/bies.201700106.
    1. Rexhaj E, Paoloni-Giacobino A, Rimoldi SF, Fuster DG, Anderegg M, Somm E, et al. Mice generated by in vitro fertilization exhibit vascular dysfunction and shortened life span. J Clin Invest. 2013;123:5052–5060. doi: 10.1172/JCI68943.
    1. Vrooman LA, Bartolomei MS. Can assisted reproductive technologies cause adult-onset disease? Evidence from human and mouse. Reprod Toxicol. 2017;68:72–84. doi: 10.1016/j.reprotox.2016.07.015.
    1. Cerny D, Sartori C, Rimoldi SF, Meister T, Soria R, Bouillet E, et al. Assisted reproductive technologies predispose to insulin resistance and obesity in male mice challenged with a high-fat diet. Endocrinology. 2017;158:1152–1159. doi: 10.1210/en.2016-1475.
    1. Cox GF, Bürger J, Lip V, Mau UA, Sperling K, Wu B-L, et al. Intracytoplasmic sperm injection may increase the risk of imprinting defects. Am J Hum Genet. 2002;71:162–164. doi: 10.1086/341096.
    1. DeBaun MR, Niemitz EL, Feinberg AP. Association of in vitro fertilization with Beckwith-Wiedemann syndrome and epigenetic alterations of LIT1 and H19. Am J Hum Genet. 2003;72:156–160. doi: 10.1086/346031.
    1. Halliday J, Oke K, Breheny S, Algar E, Amor DJ. Beckwith-Wiedemann syndrome and IVF: a case-control study. Am J Hum Genet. 2004;75:526–528. doi: 10.1086/423902.
    1. Bowdin S, Allen C, Kirby G, Brueton L, Afnan M, Barratt C, et al. A survey of assisted reproductive technology births and imprinting disorders. Hum Reprod. 2007;22:3237–3240. doi: 10.1093/humrep/dem268.
    1. Cortessis VK, Azadian M, Buxbaum J, Sanogo F, Song AY, Sriprasert I, et al. Comprehensive meta-analysis reveals association between multiple imprinting disorders and conception by assisted reproductive technology. J Assist Reprod Genet. 2018;35:943–952. doi: 10.1007/s10815-018-1173-x.
    1. Hattori H, Hiura H, Kitamura A, Miyauchi N, Kobayashi N, Takahashi S, et al. Association of four imprinting disorders and ART. Clin Epigenetics. 2019;11:1–12. doi: 10.1186/s13148-018-0606-9.
    1. Lidegaard Ø, Pinborg A, Andersen AN. Imprinting diseases and IVF: Danish national IVF cohort study. Hum Reprod. 2005;20:950–954. doi: 10.1093/humrep/deh714.
    1. Doornbos ME, Maas SM, McDonnell J, Vermeiden JPW, Hennekam RCM. Infertility, assisted reproduction technologies and imprinting disturbances: a Dutch study. Hum Reprod. 2007;22:2476–2480. doi: 10.1093/humrep/dem172.
    1. Brioude F, Kalish JM, Mussa A, Foster AC, Bliek J, Ferrero GB, et al. Clinical and molecular diagnosis, screening and management of Beckwith-Wiedemann syndrome: an international consensus statement. Nat Rev Endocrinol. 2018;14:229–249. doi: 10.1038/nrendo.2017.166.
    1. Stewart KR, Veselovska L, Kelsey G. Establishment and functions of DNA methylation in the germline. Epigenomics. 2016;8:1399–1413. doi: 10.2217/epi-2016-0056.
    1. Smallwood SA, Tomizawa SI, Krueger F, Ruf N, Carli N, Segonds-Pichon A, et al. Dynamic CpG island methylation landscape in oocytes and preimplantation embryos. Nat Genet. 2011;43:811–814. doi: 10.1038/ng.864.
    1. Gahurova L, Tomizawa S, Smallwood SA, Stewart-Morgan KR, Saadeh H, Kim J, et al. Transcription and chromatin determinants of de novo DNA methylation timing in oocytes. Epigenetics Chromatin. 2017;10:25. doi: 10.1186/s13072-017-0133-5.
    1. Veselovska L, Smallwood SA, Saadeh H, Stewart KR, Krueger F, Maupetit-Méhouas S, et al. Deep sequencing and de novo assembly of the mouse oocyte transcriptome define the contribution of transcription to the DNA methylation landscape. Genome Biol. 2015;16:209. doi: 10.1186/s13059-015-0769-z.
    1. Kobayashi H, Sakurai T, Imai M, Takahashi N, Fukuda A, Yayoi O, et al. Contribution of intragenic DNA methylation in mouse gametic DNA methylomes to establish oocyte-specific heritable marks. PLoS Genet. 2012;8:e1002440. doi: 10.1371/journal.pgen.1002440.
    1. Anckaert E, Sánchez F, Billooye K, Smitz J. Dynamics of imprinted DNA methylation and gene transcription for imprinting establishment in mouse oocytes in relation to culture duration variability. Biol Reprod. 2013;89:1–8. doi: 10.1095/biolreprod.113.111641.
    1. Anckaert E, Adriaenssens T, Romero S, Dremier S, Smitz J. Unaltered imprinting establishment of key imprinted genes in mouse oocytes after in vitro follicle culture under variable follicle-stimulating hormone exposure. Int J Dev Biol. 2009;53:541–548. doi: 10.1387/ijdb.082619ea.
    1. Anckaert E, Romero S, Adriaenssens T, Smitz J. Effects of low methyl donor levels in culture medium during mouse follicle culture on oocyte imprinting establishment. Biol Reprod. 2010;83:377–386. doi: 10.1095/biolreprod.109.082164.
    1. Heinzmann J, Hansmann T, Herrmann D, Wrenzycki C, Zechner U, Haaf T, et al. Epigenetic profile of developmentally important genes in bovine oocytes. Mol Reprod Dev. 2011;78:188–201. doi: 10.1002/mrd.21281.
    1. Kuhtz J, Romero S, De Vos M, Smitz J, Haaf T, Anckaert E. Human in vitro oocyte maturation is not associated with increased imprinting error rates at LIT1, SNRPN, PEG3 and GTL2. Hum Reprod. 2014;29:1995–2005. doi: 10.1093/humrep/deu155.
    1. Branco MR, King M, Perez-Garcia V, Bogutz AB, Caley M, Fineberg E, et al. Maternal DNA methylation regulates early trophoblast development. Dev Cell. 2016;36:152–163. doi: 10.1016/j.devcel.2015.12.027.
    1. Hanna CW, Peñaherrera MS, Saadeh H, Andrews S, DE MF, Kelsey G, et al. Pervasive polymorphic imprinted methylation in the human placenta. Genome Res. 2016;26:756–767. doi: 10.1101/gr.196139.115.
    1. Sanchez-Delgado M, Court F, Vidal E, Medrano J, Monteagudo-Sanchez A, Martin-trujillo A, et al. Human oocyte-derived methylation differences persist in the placenta revealing widespread transient imprinting. PLoS Genet. 2016;12:1–23. doi: 10.1371/journal.pgen.1006427.
    1. Saenz-de-Juano MD, Billooye K, Smitz J, Anckaert E. The loss of imprinted DNA methylation in mouse blastocysts is inflicted to a similar extent by in vitro follicle culture and ovulation induction. Mol Hum Reprod. 2016;22:427–441. doi: 10.1093/molehr/gaw013.
    1. Marshall KL, Rivera RM. The effects of superovulation and reproductive aging on the epigenome of the oocyte and embryo. Mol Reprod Dev. 2017;85(2):1–16.
    1. Market-velker BA, Zhang L, Magri LS, Bonvissuto AC, Mann MR. Dual effects of superovulation: loss of maternal and paternal imprinted methylation in a dose-dependent manner. Hum Mol Genet. 2010;19:36–51. doi: 10.1093/hmg/ddp465.
    1. Market-Velker BA, Fernandes AD, Mann MRW. Side-by-side comparison of five commercial media systems in a mouse model: suboptimal in vitro culture interferes with imprint maintenance. Biol Reprod. 2010;83:938–950. doi: 10.1095/biolreprod.110.085480.
    1. Pinter O, Beda Z, Csaba Z, Gerendai I. Differences in the onset of puberty in selected inbred mouse strains. 9th Eur Congr Endocrinol. Endocrine Abstracts: Budapest, Hungary; 2007.
    1. Cortvrindt R, Smitz J, Van Steirteghem AC. In-vitro maturation, fertilization and embryo development of immature oocytes from early preantral follicles from prepuberal mice in a simplified culture system. Hum Reprod. 1996;11:2656–2666. doi: 10.1093/oxfordjournals.humrep.a019188.
    1. Miura F, Enomoto Y, Dairiki R, Ito T. Amplification-free whole-genome bisulfite sequencing by post-bisulfite adaptor tagging. Nucleic Acids Res. 2012;40:e136. doi: 10.1093/nar/gks454.
    1. Canovas S, Ivanova E, Romar R, García-Martínez S, Soriano-Úbeda C, García-Vázquez FA, et al. DNA methylation and gene expression changes derived from assisted reproductive technologies can be decreased by reproductive fluids. Elife. 2017;6:1–24. doi: 10.7554/eLife.23670.
    1. Shirane K, Toh H, Kobayashi H, Miura F, Chiba H, Ito T, et al. Mouse oocyte methylomes at base resolution reveal genome-wide accumulation of non-CpG methylation and role of DNA Methyltransferases. PLoS Genet. 2013;9:e1003439. doi: 10.1371/journal.pgen.1003439.
    1. Wang L, Zhang J, Duan J, Gao X, Zhu W, Lu X, et al. Programming and inheritance of parental DNA methylomes in mammals. Cell. 2014;157:979–991. doi: 10.1016/j.cell.2014.04.017.
    1. Stolt CC, Lommes P, Hillgärtner S, Wegner M. The transcription factor Sox5 modulates Sox10 function during melanocyte development. Nucleic Acids Res. 2008;36:5427–5440. doi: 10.1093/nar/gkn527.
    1. Serth K, Beckers A, Schuster-Gossler K, Pavlova MN, Müller J, Paul MC, et al. Generation of an 870 kb deletion encompassing the Skt/Etl4 locus by combination of inter- and intra-chromosomal recombination. BMC Genet. 2015;16:1–17. doi: 10.1186/s12863-015-0302-0.
    1. Cameron RS, Liu C, Pihkala JPS. Myosin 16 levels fluctuate during the cell cycle and are downregulated in response to DNA replication stress. Cytoskeleton. 2013;70:328–348. doi: 10.1002/cm.21109.
    1. Horn KH, Warner DR, Pisano M, Greene RM. PRDM16 expression in the developing mouse embryo. Acta Histochem. 2011;113:150–155. doi: 10.1016/j.acthis.2009.09.006.
    1. Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature. 2008;454:961–967. doi: 10.1038/nature07182.
    1. Mochizuki N, Shimizu S, Nagasawa T, Tanaka H, Taniwaki M, Morishita K. A novel gene, MEL1, mapped to 1p36.3 is highly homologous to the MDS1/EVI1 gene and is transcriptionally activated in t(1;3)(p36;q21)-positive leukemia cells. Bone. 2010;96:3209–3214.
    1. Chen F, Lin J, Sun X, Xiao B, Ning SF, Zhu S, et al. Mechanisms by which in vitro meiotic arrest and sexual maturity improve developmental potential of mouse oocytes. Sci Rep. 2017;7:1–13. doi: 10.1038/s41598-016-0028-x.
    1. Kruse R, Krantz J, Barker N, Coletta RL, Rafikov R, Luo M, et al. Characterization of the CLASP2 protein interaction network identifies SOGA1 as a microtubule-associated protein. Mol Cell Proteomics. 2017;16:1718–1735. doi: 10.1074/mcp.RA117.000011.
    1. Flora A, Garcia JJ, Thaller C, Zoghbi HY. The E-protein Tcf4 interacts with Math1 to regulate differentiation of a specific subset of neuronal progenitors. Proc Natl Acad Sci. 2007;104:15382–15387. doi: 10.1073/pnas.0707456104.
    1. Shen S, Pu J, Lang B, McCaig CD. A zinc finger protein Zfp521 directs neural differentiation and beyond. Stem Cell Res Ther. 2011;2:2–5. doi: 10.1186/scrt61.
    1. Fleenor CJ, Arends T, Lei H, Okuyama K, Kuruvilla J, Cristobal S, et al. Zinc finger protein 521 regulates early hematopoiesis. Mol Cell Biol. 2018;38:1–16. doi: 10.1128/MCB.00603-17.
    1. Salisbury JL. Centrosomes: Sfi1p and centrin unravel a structural riddle. Curr Biol. 2004;14:27–29. doi: 10.1016/j.cub.2003.12.019.
    1. Weber M, Guibert S, Forne T. Global profiling of DNA methylation erasure in mouse primordial germ cells. Genome Res. 2012;22:633–641. doi: 10.1101/gr.130997.111.
    1. Kraus P, Sivakamasundari V, Yu HB, Xing X, Lim SL, Adler T, et al. Pleiotropic functions for transcription factor Zscan10. PLoS One. 2014;9:1–14.
    1. Zhang W, Walker E, Tamplin OJ, Rossant J, Stanford WL, Hughes TR. Zfp206 regulates ES cell gene expression and differentiation. Nucleic Acids Res. 2006;34:4780–4790. doi: 10.1093/nar/gkl631.
    1. Kawashima H, Sugito K, Yoshizawa S, Uekusa S, Furuya T, Ikeda T, et al. DNA hypomethylation at the ZNF206-exon 5 CpG island associated with neuronal differentiation in mice and development of neuroblastoma in humans. Int J Oncol. 2012;40:31–9.
    1. Kurowski A, Molotkov A, Soriano P. FGFR1 regulates trophectoderm development and facilitates blastocyst implantation. Dev Biol. 2019;446(1):94–101. doi: 10.1016/j.ydbio.2018.12.008.
    1. Li H, Liefke R, Jiang J, Kurland JV, Tian W, Deng P, et al. Polycomb-like proteins link the PRC2 complex to CpG islands. Nature. 2017;549(7671):287–291. doi: 10.1038/nature23881.
    1. Ma J, Fukuda Y, Schultz RM. Mobilization of dormant Cnot7 mRNA promotes deadenylation of maternal transcripts during mouse oocyte maturation. Biol Reprod. 2015;93(2):48. doi: 10.1095/biolreprod.115.130344.
    1. Pan H, Brien MJO, Wigglesworth K, Eppig JJ, Schultz RM. Transcript profiling during mouse oocyte development and the effect of gonadotropin priming and development in vitro. Dev Biol. 2005;286:493–506. doi: 10.1016/j.ydbio.2005.08.023.
    1. Denomme MM, Zhang L, Mann MRW. Embryonic imprinting perturbations do not originate from superovulation-induced defects in DNA methylation acquisition. Fertil Steril. 2011;96:734–738. doi: 10.1016/j.fertnstert.2011.06.055.
    1. Han L, Ren C, Li L, Li X, Ge J, Wang H, et al. Embryonic defects induced by maternal obesity in mice derive from Stella insufficiency in oocytes. Nat Genet. 2018;50:432–442. doi: 10.1038/s41588-018-0055-6.
    1. Fonseca-Balvís N, Garcia-Martinez S, Pérez-Cerezales S, Ivanova E, Gomez-Redondo I, Hamdi M, et al. Cultured bovine embryo biopsy conserves methylation marks from original embryo. Biol Reprod. 2017;97:189–196. doi: 10.1093/biolre/iox077.
    1. Stewart KR, Veselovska L, Kim J, Huang J, Saadeh H, Tomizawa S, et al. Dynamic changes in histone modifications precede de novo DNA methylation in oocytes. Genes Dev. 2015;29(23):2449–2462. doi: 10.1101/gad.271353.115.
    1. Morris JH, Huerta-Cepas J, Junge A, Szklarczyk D, Jensen LJ, von Mering C, et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2018;47:D607–D613.

Source: PubMed

3
Tilaa