Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Embryonic defects induced by maternal obesity in mice derive from Stella insufficiency in oocytes

A Publisher Correction to this article was published on 27 February 2018

This article has been updated

Abstract

Maternal obesity can impair embryo development and offspring health, yet the mechanisms responsible remain poorly understood. In a high-fat diet (HFD)-based female mouse model of obesity, we identified a marked reduction of Stella (also known as DPPA3 or PGC7) protein in oocytes. Starting with this clue, we found that the establishment of pronuclear epigenetic asymmetry in zygotes from obese mice was severely disrupted, inducing the accumulation of maternal 5-hydroxymethylcytosine modifications and DNA lesions. Furthermore, methylome-wide sequencing analysis detected global hypomethylation across the zygote genome in HFD-fed mice, with a specific enrichment in transposon elements and unique regions. Notably, overexpression of Stella in the oocytes of HFD-fed mice not only restored the epigenetic remodeling in zygotes but also partly ameliorated the maternal-obesity-associated developmental defects in early embryos and fetal growth. Thus, Stella insufficiency in oocytes may represent a critical mechanism that mediates the phenotypic effects of maternal obesity in embryos and offspring.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Developmental phenotypes of preimplantation and postimplantation embryos from HFD mice.
Fig. 2: Reduced accumulation of Stella protein in MII oocytes from obese mice.
Fig. 3: Rapid loss of 5mC from the maternal pronucleus of zygotes from HFD mice.
Fig. 4: Maternal TET3-dependent 5hmC and γ-H2AX accumulation in HFD zygotes contributes to early embryonic developmental defects.
Fig. 5: Global DNA hypomethylation across different genomic features in zygotes from HFD mice.
Fig. 6: Nonrandom distribution of regions differentially methylated between genomes from ND and HFD zygotes.
Fig. 7: Effects of Stella overexpression on methylation status and developmental progression of embryos generated by IVF of oocytes obtained from HFD mothers.
Fig. 8: Diagram illustrating the proposed mechanisms for alteration of embryo development and offspring phenotype by maternal obesity.

Similar content being viewed by others

Change history

References

  1. King, J. C. Maternal obesity, metabolism and pregnancy outcomes. Annu. Rev. Nutr. 26, 271–291 (2006).

    Article  PubMed  CAS  Google Scholar 

  2. Tenenbaum-Gavish, K. & Hod, M. Impact of maternal obesity on fetal health. Fetal Diagn. Ther. 34, 1–7 (2013).

    Article  PubMed  Google Scholar 

  3. Neri, C. & Edlow, A. G. Effects of maternal obesity on fetal programming: molecular approaches. Cold Spring Harb. Perspect. Med. 6, a026591 (2015).

    Article  PubMed  CAS  Google Scholar 

  4. Mahizir, D. et al. Maternal obesity in females born small: pregnancy complications and offspring disease risk. Mol. Nutr. Food Res. 60, 8–17 (2016).

    Article  PubMed  CAS  Google Scholar 

  5. Luzzo, K. M. et al. High-fat-diet-induced developmental defects in the mouse: oocyte meiotic aneuploidy and fetal growth retardation and brain defects. PLoS One 7, e49217 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. McPherson, N. O. et al. When two obese parents are worse than one! Impacts on embryo and fetal development. Am. J. Physiol. Endocrinol. Metab. 309, E568–E581 (2015).

    Article  PubMed  CAS  Google Scholar 

  7. Wu, L. L. et al. Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 142, 681–691 (2015).

    Article  PubMed  CAS  Google Scholar 

  8. Jungheim, E. S. et al. Diet-induced obesity model: abnormal oocytes and persistent growth abnormalities in the offspring. Endocrinology 151, 4039–4046 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Sasson, I. E., Vitins, A. P., Mainigi, M. A., Moley, K. H. & Simmons, R. A. Pregestational versus gestational exposure to maternal obesity differentially programs the offspring in mice. Diabetologia 58, 615–624 (2015).

    Article  PubMed  CAS  Google Scholar 

  10. Finger, B. J., Harvey, A. J., Green, M. P. & Gardner, D. K. Combined parental obesity negatively impacts preimplantation mouse embryo development, kinetics, morphology and metabolism. Hum. Reprod. 30, 2084–2096 (2015).

    Article  PubMed  CAS  Google Scholar 

  11. Huypens, P. et al. Epigenetic germline inheritance of diet-induced obesity and insulin resistance. Nat. Genet. 48, 497–499 (2016).

    Article  PubMed  CAS  Google Scholar 

  12. Wyman, A., Pinto, A. B., Sheridan, R. & Moley, K. H. One-cell zygote transfer from diabetic to nondiabetic mouse results in congenital malformations and growth retardation in offspring. Endocrinology 149, 466–469 (2008).

    Article  PubMed  CAS  Google Scholar 

  13. Grindler, N. M. & Moley, K. H. Maternal obesity, infertility and mitochondrial dysfunction: potential mechanisms emerging from mouse model systems. Mol. Hum. Reprod. 19, 486–494 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Igosheva, N. et al. Maternal-diet-induced obesity alters mitochondrial activity and redox status in mouse oocytes and zygotes. PLoS One 5, e10074 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Hou, Y. J. et al. Both diet- and gene-mutation-induced obesity affect oocyte quality in mice. Sci. Rep. 6, 18858 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Zhang, L. et al. Sirt3 prevents maternal-obesity-associated oxidative stress and meiotic defects in mouse oocytes. Cell Cycle 14, 2959–2968 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Saben, J. L. et al. Maternal metabolic syndrome programs mitochondrial dysfunction via germline changes across three generations. Cell Rep. 16, 1–8 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Nakamura, T. et al. PGC7 (Stella) protects against DNA demethylation in early embryogenesis. Nat. Cell Biol. 9, 64–71 (2007).

    Article  PubMed  CAS  Google Scholar 

  19. Bermejo-Alvarez, P., Rosenfeld, C. S. & Roberts, R. M. Effect of maternal obesity on estrous cyclicity, embryo development and blastocyst gene expression in a mouse model. Hum. Reprod. 27, 3513–3522 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Payer, B. et al. Stella is a maternal effect gene required for normal early development in mice. Curr. Biol. 13, 2110–2117 (2003).

    Article  PubMed  CAS  Google Scholar 

  21. Bortvin, A., Goodheart, M., Liao, M. & Page, D. C. Dppa3 (Pgc7 or Stella) is a maternal factor and is not required for germ cell specification in mice. BMC Dev. Biol. 4, 2 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Funaki, S. et al. Inhibition of maintenance DNA methylation by Stella. Biochem. Biophys. Res. Commun. 453, 455–460 (2014).

    Article  PubMed  CAS  Google Scholar 

  23. Fernandez-Twinn, D. S., Constância, M. & Ozanne, S. E. Intergenerational epigenetic inheritance in models of developmental programming of adult disease. Semin. Cell Dev. Biol. 43, 85–95 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Iqbal, K., Jin, S. G., Pfeifer, G. P. & Szabó, P. E. Reprogramming of the paternal genome upon fertilization involves genome-wide oxidation of 5-methylcytosine. Proc. Natl. Acad. Sci. USA 108, 3642–3647 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Gu, T. P. et al. The role of Tet3 DNA dioxygenase in epigenetic reprogramming by oocytes. Nature 477, 606–610 (2011).

    Article  PubMed  CAS  Google Scholar 

  26. Yu, C. et al. CRL4 complex regulates mammalian oocyte survival and reprogramming by activation of Tet proteins. Science 342, 1518–1521 (2013).

    Article  PubMed  CAS  Google Scholar 

  27. Amouroux, R. et al. De novo DNA methylation drives 5hmC accumulation in mouse zygotes. Nat. Cell Biol. 18, 225–233 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Kang, J., Kalantry, S. & Rao, A. PGC7, H3K9me2 and Tet3: regulators of DNA methylation in zygotes. Cell Res. 23, 6–9 (2013).

    Article  PubMed  CAS  Google Scholar 

  29. Nakamura, T. et al. PGC7 binds histone H3K9me2 to protect against conversion of 5mC to 5hmC in early embryos. Nature 486, 415–419 (2012).

    Article  PubMed  CAS  Google Scholar 

  30. Bian, C. & Yu, X. PGC7 suppresses Tet3 for protecting DNA methylation. Nucleic Acids Res. 42, 2893–2905 (2014).

    Article  PubMed  CAS  Google Scholar 

  31. Kafer, G. R. et al. 5-hydroxymethylcytosine marks sites of DNA damage and promotes genome stability. Cell Rep. 14, 1283–1292 (2016).

    Article  PubMed  CAS  Google Scholar 

  32. Ladstatter, S. & Tachibana-Konwalski, K. A surveillance mechanism ensures repair of DNA lesions during zygotic reprogramming. Cell 167, 1774–1787 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Wossidlo, M. et al. Dynamic link of DNA demethylation, DNA strand breaks and repair in mouse zygotes. EMBO J. 29, 1877–1888 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Nakatani, T. et al. Stella preserves maternal chromosome integrity by inhibiting 5hmC-induced γ-H2AX accumulation. EMBO Rep. 16, 582–589 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Tsukada, Y., Akiyama, T. & Nakayama, K. I. Maternal Tet3 is dispensable for embryonic development but is required for neonatal growth. Sci. Rep. 5, 15876 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Inoue, A., Shen, L., Matoba, S. & Zhang, Y. Haploinsufficiency, but not defective paternal 5mC oxidation, accounts for the developmental defects of maternal Tet3 knockouts. Cell Rep. 10, 463–470 (2015).

    Article  PubMed  CAS  Google Scholar 

  37. Smallwood, S. A. et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 11, 817–820 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Huang, Y. et al. The behavior of 5-hydroxymethylcytosine in bisulfite sequencing. PLoS One 5, e8888 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Bestor, T. H. & Bourc’his, D. Transposon silencing and imprint establishment in mammalian germ cells. Cold Spring Harb. Symp. Quant. Biol. 69, 381–387 (2004).

    Article  PubMed  CAS  Google Scholar 

  40. Anderson, N. H., Sadler, L. C., Stewart, A. W., Fyfe, E. M. & McCowan, L. M. Independent risk factors for infants who are small for gestational age by customized birthweight centiles in a multi-ethnic New Zealand population. Aust. N. Z. J. Obstet. Gynaecol. 53, 136–142 (2013).

    Article  PubMed  Google Scholar 

  41. Leddy, M. A., Power, M. L. & Schulkin, J. The impact of maternal obesity on maternal and fetal health. Rev. Obstet. Gynecol. 1, 170–178 (2008).

    PubMed  PubMed Central  Google Scholar 

  42. Jones, H. N. et al. High-fat diet before and during pregnancy causes marked upregulation of placental nutrient transport and fetal overgrowth in C57/BL6 mice. FASEB J. 23, 271–278 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Aye, I. L., Rosario, F. J., Powell, T. L. & Jansson, T. Adiponectin supplementation in pregnant mice prevents the adverse effects of maternal obesity on placental function and fetal growth. Proc. Natl Acad. Sci. USA 112, 12858–12863 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Rando, O. J. & Simmons, R. A. I’m eating for two: parental dietary effects on offspring metabolism. Cell 161, 93–105 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Luke, B. et al. The effect of increasing obesity on the response to and outcome of assisted reproductive technology: a national study. Fertil. Steril. 96, 820–825 (2011).

    Article  PubMed  Google Scholar 

  46. Sato, M. et al. Identification of PGC7, a new gene expressed specifically in preimplantation embryos and germ cells. Mech. Dev. 113, 91–94 (2002).

    Article  PubMed  CAS  Google Scholar 

  47. Huang, Y. et al. Stella modulates transcriptional and endogenous retrovirus programs during maternal-to-zygotic transition. eLife 6, e22345 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Capy, P., Gasperi, G., Biémont, C. & Bazin, C. Stress and transposable elements: co-evolution or useful parasites? Heredity 85, 101–106 (2000).

    Article  PubMed  CAS  Google Scholar 

  49. Faulk, C. et al. Detection of differential DNA methylation in repetitive DNA of mice and humans perinatally exposed to bisphenol A. Epigenetics 11, 489–500 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Sun, M. et al. 5-hydroxymethylcytosine-mediated alteration of transposon activity associated with the exposure to adverse in utero environments in human. Hum. Mol. Genet. 25, 2208–2219 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Itoh, M. et al. Role of Gab1 in heart, placenta and skin development, and growth-factor- and cytokine-induced extracellular-signal-regulated kinase mitogen-activated protein kinase activation. Mol. Cell. Biol. 20, 3695–3704 (2000).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Kornfeld, J. W. et al. Obesity-induced overexpression of miR-802 impairs glucose metabolism through silencing of Hnf1b. Nature 494, 111–115 (2013).

    Article  PubMed  CAS  Google Scholar 

  53. Santos, F., Hendrich, B., Reik, W. & Dean, W. Dynamic reprogramming of DNA methylation in the early mouse embryo. Dev. Biol. 241, 172–182 (2002).

    Article  PubMed  CAS  Google Scholar 

  54. Adenot, P. G., Mercier, Y., Renard, J. P. & Thompson, E. M. Differential H4 acetylation of paternal and maternal chromatin precedes DNA replication and differential transcriptional activity in pronuclei of 1-cell mouse embryos. Development 124, 4615–4625 (1997).

    PubMed  CAS  Google Scholar 

  55. Nagy, A. Manipulating the Mouse Embryo: A Laboratory Manual (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2003).

  56. Hou, X. et al. Differing roles of pyruvate dehydrogenase kinases during mouse oocyte maturation. J. Cell Sci. 128, 2319–2329 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Qi, L. et al. Systematic analysis of the phosphoproteome and kinase–substrate networks in the mouse testis. Mol. Cell. Proteomics 13, 3626–3638 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Wang, J. et al. Quantitative phosphoproteomics analysis reveals a key role of insulin growth factor 1 receptor (IGF1R) tyrosine kinase in human sperm capacitation. Mol. Cell. Proteomics 14, 1104–1112 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Ge, Z. J. et al. DNA methylation in oocytes and liver of female mice and their offspring: effects of high-fat-diet-induced obesity. Environ. Health Perspect. 122, 159–164 (2014).

    Article  PubMed  Google Scholar 

  60. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-seq applications. Bioinformatics 27, 1571–1572 (2011).

    Google Scholar 

  61. Speir, M. L. et al. The UCSC Genome Browser database: 2016 update. Nucleic Acids Res. 44 (D1), D717–D725 (2016).

    Article  PubMed  CAS  Google Scholar 

  62. O’Leary, N. A. et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion and functional annotation. Nucleic Acids Res. 44 (D1), D733–D745 (2016).

    Article  PubMed  CAS  Google Scholar 

  63. Jühling, F. et al. metilene: fast and sensitive calling of differentially methylated regions from bisulfite sequencing data. Genome Res. 26, 256–262 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Huang, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to T. Schedl (Washington University in St. Louis School of Medicine) for a valuable discussion that led to the experiment design reported in this paper and his critical reading of the manuscript. We also thank Q.-Y. Sun and Z.-B. Wang (Chinese Academy of Sciences) for providing plasmids and Y.-S. Guo (Nanjing Medical University) for help with the proteomic assay. This work was supported by National Key Scientific Research Projects of China (2014CB943200 to Q.W.), the National Key Research and Development Program of China (2017YFC1001500 to Q.W.), the National Natural Science Foundation of China (no. 31571543 to Q.W.), a Major Research Plan of the National Natural Science Foundation of China (no. U1435222 to W.S.), a Major Research Plan of the National Key R&D Program of China (no. 2016YFC0901600 to W.S.), and the National High Technology Research and Development Program of China (no. 2015AA020108 to W.S.).

Author information

Authors and Affiliations

Authors

Contributions

Q.W., W.S. and K.H.M. conceived the project; X.G. contributed to the proteomic experiment; L.H., L.L., X.L. and H.W. performed the experiments on embryo staining and methylation validation; C.R. and W.S. contributed to the sequencing and bioinformatics analysis; L.H., L.L., J.G. and Y.M. performed IVF, microinjection and embryo transfer experiments; and Q.W. and W.S. wrote and K.H.M. revised the manuscript.

Corresponding authors

Correspondence to Wenjie Shu or Qiang Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 High-fat diet leads to obesity, glucose intolerance and insulin resistance in female mice.

a, Female mice receiving a normal diet (ND) or high-fat diet (HFD) for 16 weeks. bf, Mice were assessed for body fat (b), body weight (c), blood glucose (d) and serum insulin (e) during GTT, and blood glucose during ITT (f) (n = 10 for ND mice; n = 12 for HFD mice). Data are expressed as mean ± s.d. Student’s t test (two-tailed) was used for statistical analysis.

Supplementary Figure 2 Effects of maternal HFD on ovulation and litter size in mice.

a, Maternal HFD did not significantly affect the average number of ovulated eggs in mice, although there was a slight reduction (n = 8 for ND mice; n = 8 for HFD mice). b, Average litter size was significantly reduced in HFD mice compared to ND mice when these mice were mated with normal males (n = 8 for ND mice; n = 8 for HFD mice). Student’s t test (two-tailed) was used for statistical analysis. Error bars, s.d.; center values, mean; n.s., not significant.

Supplementary Figure 3 Female ob/ob mice exhibit obesity, glucose intolerance and insulin resistance.

a, Eight-week-old female wild-type (WT) and ob/ob mice (n = 8 for WT mice; n = 10 for ob/ob mice). be, Mice were assessed for body fat (b), body weight (c), and blood glucose (d) and serum insulin (e) during GTT. Student’s t test (two-tailed) was used for statistical analysis. Error bars, s.d.; center values, mean; n.s., not significant.

Supplementary Figure 4 Maternal obesity does not significantly affect Stella mRNA abundance in oocytes.

Relative quantification of Stella mRNA levels in MII oocytes from ND and HFD mice or WT and ob/ob mice was conducted by real-time RT–PCR. Data are expressed as means ± s.d. from three independent experiments. Student’s t test (two-tailed) was used for statistical analysis. Center values, mean; n.s., not significant.

Supplementary Figure 5 Rapid loss of 5mC in the maternal genome of zygotes from ob/ob mice.

a, Representative images of zygotes from wild-type (WT) and ob/ob mice stained with anti-5mC (red) and anti-5hmC (green) antibodies. ♂ and ♀ indicate the paternal pronucleus and maternal pronucleus, respectively. Arrowheads indicate 5mC loss and 5hmC accumulation in the maternal pronuclei of ob/ob zygotes. PB, polar body. Scale bars, 25 μm. b,c, Quantification of the relative levels of 5mC and 5hmC in both pronuclei of zygotes. Results are presented as signal intensity in paternal and maternal pronuclei (left axis) or a ratio of paternal over maternal signal (right axis). Each data point represents a zygote (n = 12 biologically independent zygotes for each group). Student’s t test (two-tailed) was used for statistical analysis. Error bars, s.d.; center values, mean; n.s., not significant.

Supplementary Figure 6 Maternal HFD has no effects on the intensity of 5mC staining in mouse oocytes.

a, Representative images of MII oocytes from ND and HFD mice stained with anti-5mC antibody (5mC, green). DNA was counterstained with propidium iodide (red). PB, polar body. Scale bars, 25 μm. b, Quantification of the relative levels of 5mC in oocytes. Each data point represents an oocyte (n = 14 biologically independent oocytes for each group). Student’s t test (two-tailed) was used for statistical analysis. Error bars, s.d.; center values, mean; n.s., not significant.

Supplementary Figure 7 Methylation status of histone H3K9me2 in oocytes and zygotes from ND and HFD mice.

a, Representative images of MII oocytes from ND and HFD mice stained with H3K9me2 antibody (H3K9me2; green). DNA was counterstained with propidium iodide (red). b, Representative images of zygotes from ND and HFD mice stained with H3K9me2 antibody (H3K9me2, green; DNA, red). ♂ and ♀ indicate the paternal pronucleus and maternal pronucleus, respectively. PB, polar body. Scale bars, 25 μm. c,d, Quantification of the H3K9me2 fluorescence in MII oocytes (n = 12 biologically independent oocytes for each group) and the maternal pronuclei of zygotes (n = 12 biologically independent zygotes for each group), with the ND values set to one. Student’s t test (two-tailed) was used for statistical analysis. Error bars, s.d.; center values, mean; n.s., not significant.

Supplementary Figure 8 TET3 expression in oocytes from ND and HFD mice.

a, The relative levels of Tet3 mRNA in MII oocytes obtained from ND and HFD mice were determined by real-time RT–PCR, with Gapdh as an internal control. Data are expressed as the means ± s.d. from three independent experiments. Student’s t test (two-tailed) was used for statistical analysis. Center values, mean; n.s., not significant. b, TET3 protein levels in MII oocytes from ND and HFD mice were evaluated by western blot, with actin as a loading control. The experiments were repeated three times independently with similar results. Band intensity was calculated using ImageJ software, and the ratio of TET3/actin expression was normalized and values are indicated.

Supplementary Figure 9 Abnormal γ-H2AX accumulation in the maternal pronuclei of zygotes from HFD mice.

a, Representative images of γ-H2AX (green) and nuclear (red) staining on ND zygotes, HFD zygotes, HFD zygotes injected with PBS or Stella mRNA. ♂ and ♀ indicate the paternal pronucleus and maternal pronucleus, respectively. Arrows indicate the apparent γ-H2AX accumulation in the maternal pronuclei of HFD zygotes. PB, polar body. Scale bars, 25 μm. b, Quantification of the numbers of γ-H2AX foci in maternal and paternal pronuclei. Each data point represents a zygote (n = 11 for ND, n = 10 for HFD, n = 11 for HFD + PBS, and n = 12 for HFD + Stella, biologically independent zygotes). Statistical analyses were performed with one-way ANOVA with Tukey’s post hoc test. Adjustments were made for multiple comparisons. Error bars, s.d.; center values, mean; n.s., not significant.

Supplementary Figure 10 Knockdown of endogenous TET3 protein in MII oocytes.

Specifically designed siRNAs for TET3 were microinjected into MII oocytes. Samples (100 oocytes per lane) were processed for western blot analysis; actin was served as a loading control. The experiments were repeated three times independently with similar results. Band intensity was calculated using ImageJ software, and the ratio of TET3/actin expression was normalized and values are indicated. Images have been cropped; please compare to Supplementary Data.

Supplementary Figure 11 TET3-dependent 5hmc and γ-H2AX accumulation in HFD zygotes contributes to early embryonic developmental defects.

a, Representative images of 5mC (red) and 5hmC (green) staining on ND zygotes, ND + DMOG zygotes, HFD zygotes and HFD + DMOG zygotes. b,c, Quantification of the levels of 5hmC and 5mC signal in the paternal and maternal pronuclei of zygote (n = 10 biologically independent zygotes for each group). d, Representative images of γ-H2AX (green) and nuclear (red) staining on ND zygotes, ND + DMOG zygotes, HFD zygotes and HFD + DMOG zygotes. e, Quantification of the numbers of γ-H2AX foci in maternal and paternal pronuclei. ♂ and ♀ indicate the paternal and maternal pronucleus, respectively. PB, polar body. Scale bars, 25 μm. Each data point represents a zygote (n = 12 biologically independent zygotes for each group). f, The percentage of zygotes that successfully progressed to the blastocyst stage during in vitro culture. Data are presented as the means ± s.d. from three independent experiments (total number of embryos analyzed: n = 90 for ND, n = 112 for ND + DMOG, n = 95 for HFD and n = 92 for HFD + DMOG). In e and f, statistical analyses were performed with one-way ANOVA with Tukey’s post hoc test. Adjustments were made for multiple comparisons. In all other panels, Student’s t test (two-tailed) was used for statistical analysis. Error bars, s.d.; center values, mean; n.s., not significant.

Supplementary Figure 12 Global DNA hypomethylation across different genomic features in zygotes from HFD mice.

Violin plots showing the methylation levels of the major repetitive elements in zygotes from ND and HFD mice. Mean methylation levels are indicated by the numerical value and green cross. Statistical analyses were performed by bootstrap test.

Supplementary Figure 13 Heat map analysis of gene promoters differentially methylated between ND and HFD zygotes.

DNA methylation level is colored from yellow to red to indicate low to high. The right panels show the enriched GO terms of biological processes.

Supplementary Figure 14 Overexpression of exogenous Stella protein in MII oocytes.

PBS (control group) or exogenous Myc-Stella mRNA (overexpression group) was microinjected into MII oocytes. Oocyte samples were processed for western blot analysis (100 oocytes per lane), probing with anti-Myc and anti-Stella antibody, respectively. Tubulin served as a loading control. Images have been cropped; please compare to Supplementary Data. The experiments were repeated three times independently with similar results.

Supplementary Figure 15 Effects of Stella overexpression on the methylation status of zygotes in HFD mice.

a,c,e,g, Graphical representation of the methylation pattern at the example loci of RLTR10-int, Gab1, Gstp1 and Park2 in ND (top, blue) and HFD (bottom, red) zygotes. The regions highlighted by green boxes were chosen for further validation. b,d,f,h, Bisulfite sequencing for verifying the methylation status of RLTR10-int, Gab1, Gstp1 and Park2 in ND zygotes, HFD zygotes, and HFD zygotes injected with PBS or Stella mRNA. Open and filled circles represent unmethylated and methylated CpGs, respectively. The percentages of methylated CpGs are shown below each panel. The experiments were repeated three times independently with similar results.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 and Supplementary Tables 1–6

Life Sciences Reporting Summary

Supplementary Data

Uncropped western blot images

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Han, L., Ren, C., Li, L. et al. Embryonic defects induced by maternal obesity in mice derive from Stella insufficiency in oocytes. Nat Genet 50, 432–442 (2018). https://doi.org/10.1038/s41588-018-0055-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0055-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing